54
Chapter 16 DNA Damage, DNA Repair and Cancer Carol Bernstein, Anil R. Prasad, Valentine Nfonsam and Harris Bernstein Additional information is available at the end of the chapter http://dx.doi.org/10.5772/53919 1. Introduction DNA damage appears to be a fundamental problem for life. In this chapter we review evi‐ dence indicating that DNA damages are a major primary cause of cancer. DNA damages give rise to mutations and epimutations that, by a process of natural selection, can cause progression to cancer. First, we describe the distinguishing characteristics of DNA damage, mutation and epimutation. DNA damage is a change in the basic structure of DNA that is not itself replicated when the DNA is replicated. A DNA damage can be a chemical addition or disruption to a base of DNA (creating an abnormal nucleotide or nucleotide fragment) or a break in one or both chains of the DNA strands. When DNA carrying a damaged base is replicated, an incorrect base can often be inserted opposite the site of the damaged base in the complementary strand, and this can become a mutation in the next round of replication. Also DNA double- strand breaks may be repaired by an inaccurate repair process leading to mutations. In addi‐ tion, a double strand break can cause rearrangements of the chromosome structure (possibly disrupting a gene, or causing a gene to come under abnormal regulatory control), and, if such a change can be passed to successive cell generations, it is also a form of mutation. Mu‐ tations, however, can be avoided if accurate DNA repair systems recognize DNA damages as abnormal structures, and repair the damages prior to replication. As illustrated in Figure 1, when DNA damages occur, DNA repair is a crucial protective process blocking entry of cells into carcinogenesis. We note that DNA damages occur in both replicating, proliferative cells (e.g. those forming the internal lining of the colon or blood forming “hematopoietic” cells), and in differentiat‐ ed, non-dividing cells (e.g. neurons in the brain or myocytes in muscle). Cancers occur pri‐ marily in proliferative tissues. If DNA damages in proliferating cells are not repaired due to © 2013 Bernstein et al.; licensee InTech. This is an open access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/3.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

DNA Damage, DNA Repair and Cancer - InTech - Opencdn.intechopen.com/pdfs/43929/InTech-Dna_damage... · Chapter 16 DNA Damage, DNA Repair and Cancer Carol Bernstein, Anil R. Prasad,

Embed Size (px)

Citation preview

Page 1: DNA Damage, DNA Repair and Cancer - InTech - Opencdn.intechopen.com/pdfs/43929/InTech-Dna_damage... · Chapter 16 DNA Damage, DNA Repair and Cancer Carol Bernstein, Anil R. Prasad,

Chapter 16

DNA Damage, DNA Repair and Cancer

Carol Bernstein, Anil R. Prasad,Valentine Nfonsam and Harris Bernstein

Additional information is available at the end of the chapter

http://dx.doi.org/10.5772/53919

1. Introduction

DNA damage appears to be a fundamental problem for life. In this chapter we review evi‐dence indicating that DNA damages are a major primary cause of cancer. DNA damagesgive rise to mutations and epimutations that, by a process of natural selection, can causeprogression to cancer. First, we describe the distinguishing characteristics of DNA damage,mutation and epimutation.

DNA damage is a change in the basic structure of DNA that is not itself replicated when theDNA is replicated. A DNA damage can be a chemical addition or disruption to a base ofDNA (creating an abnormal nucleotide or nucleotide fragment) or a break in one or bothchains of the DNA strands. When DNA carrying a damaged base is replicated, an incorrectbase can often be inserted opposite the site of the damaged base in the complementarystrand, and this can become a mutation in the next round of replication. Also DNA double-strand breaks may be repaired by an inaccurate repair process leading to mutations. In addi‐tion, a double strand break can cause rearrangements of the chromosome structure (possiblydisrupting a gene, or causing a gene to come under abnormal regulatory control), and, ifsuch a change can be passed to successive cell generations, it is also a form of mutation. Mu‐tations, however, can be avoided if accurate DNA repair systems recognize DNA damagesas abnormal structures, and repair the damages prior to replication. As illustrated in Figure1, when DNA damages occur, DNA repair is a crucial protective process blocking entry ofcells into carcinogenesis.

We note that DNA damages occur in both replicating, proliferative cells (e.g. those formingthe internal lining of the colon or blood forming “hematopoietic” cells), and in differentiat‐ed, non-dividing cells (e.g. neurons in the brain or myocytes in muscle). Cancers occur pri‐marily in proliferative tissues. If DNA damages in proliferating cells are not repaired due to

© 2013 Bernstein et al.; licensee InTech. This is an open access article distributed under the terms of theCreative Commons Attribution License (http://creativecommons.org/licenses/by/3.0), which permitsunrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Page 2: DNA Damage, DNA Repair and Cancer - InTech - Opencdn.intechopen.com/pdfs/43929/InTech-Dna_damage... · Chapter 16 DNA Damage, DNA Repair and Cancer Carol Bernstein, Anil R. Prasad,

inadequate expression of a DNA repair gene, this increases the risk of cancer. In contrast,when DNA damages occur in non-proliferating cells and are not repaired due to inadequateexpression of a DNA repair gene, the damages can accumulate and cause premature aging.As examples, deficiencies in DNA repair genes ERCC1 or XPF [1] or in WRN [2, 3] causeboth increased risk of cancer as well as premature aging. In Figure 1, DNA repair is indicat‐ed as a crucial process impeding both cancer and premature aging.

Figure 1. The roles of DNA damage and DNA repair in cancer and aging.

New Research Directions in DNA Repair414

Page 3: DNA Damage, DNA Repair and Cancer - InTech - Opencdn.intechopen.com/pdfs/43929/InTech-Dna_damage... · Chapter 16 DNA Damage, DNA Repair and Cancer Carol Bernstein, Anil R. Prasad,

A mutation is a change in the DNA sequence in which normal base pairs are substituted,added, deleted or rearranged. The DNA containing a mutation still consists of a sequence ofstandard base pairs, and the altered DNA sequence can be copied when the DNA is replicat‐ed. A mutation can prevent a gene from carrying out its function, or it can cause a gene to betranslated into a protein that functions abnormally. Mutations can activate oncogenes, inac‐tivate tumor suppressor genes or cause genomic instability in replicating cells, and an as‐semblage of such mutations, together in the same cell, can lead to cancer. Cancers usuallyarise from an assemblage of mutations conferring a selective advantage that leads to clonalexpansion (Figure 1). Colon cancers, for example, have an average of 15 “driver” mutations(mutations occurring repeatedly in different colon cancers) and about 75 “passenger” muta‐tions (mutations occurring infrequently in colon cancers) [4, 5]. Colon cancers also werefound to have an average of 9 duplications or deletions of chromosome segments [6] or,more recently, 17 focal amplifications, 28 recurrent deletions and up to 10 translocations [5].Since mutations have normal DNA structure, they cannot be recognized or removed byDNA repair processes in living cells. Removal of a mutation only occurs if it is sufficientlydeleterious to cause the death of the cell.

Another type of inheritable alteration, similar in some ways to a mutation, is an epige‐netic change. An epigenetic change refers to a functionally relevant modification of theDNA, or of the histone proteins controlling the relaxation or tightened winding of theDNA within their nucleosome structures. Some epigenetic changes involve specific altera‐tions of the DNA nucleotides. Examples of such changes include methylation of theDNA at particular sites (CpG islands) where the DNA starts to be transcribed into RNA.These changes may inhibit transcription. Other epigenetic changes involve modificationof histones associated with particular regions of the DNA. These may inhibit or promotethe ability of these regions to be transcribed into mRNA. Methylation of CpG islands ormodification of histones can directly alter transcription of gene-encoded mRNAs butthey can also occur in parts of the genome that code for microRNAs (miRNAs). MiRNAsare endogenous short non-protein coding RNAs (~22 nucleotides long) that post-tran‐scriptionally regulate mRNA expression in a sequence specific manner. miRNAs eithercause degradation of mRNAs or block their translation. Epigenetic modifications canplay a role similar to mutation in carcinogenesis, and about 280 cancer prone epigeneticalterations are listed by Schnekenburger and Diederich [7]. Epigenetic alterations are usu‐ally copied onto the daughter chromosomes when the parental chromosome replicates.

Although epigenetic changes can be passed down from one cell generation to the next,they are not regarded as true mutations. Most epigenetic changes appear to be part ofthe differentiation program of the cell and are necessary to allow different types of cellsto carry out different functions. In most cells of a human body, only about 5% of genesare active at any one time, often due to epigenetic modifications. However, abnormal un‐programmed epigenetic changes may also occur that alter the functioning of a cell andthese changes are referred to as “epimutations.” Programmed epigenetic changes can bereversed. During development, as daughter cells of a stem cell differentiate, some epige‐

DNA Damage, DNA Repair and Cancerhttp://dx.doi.org/10.5772/53919

415

Page 4: DNA Damage, DNA Repair and Cancer - InTech - Opencdn.intechopen.com/pdfs/43929/InTech-Dna_damage... · Chapter 16 DNA Damage, DNA Repair and Cancer Carol Bernstein, Anil R. Prasad,

netic changes are programmed for reversal. However, a double strand break in DNA (atype of DNA damage) can initiate unprogrammed epigenetic gene silencing both bycausing methylation of a CpG island as well as by promoting silencing types of histonemodifications [8]. Another form of epigenetic silencing may occur during DNA repair.The enzyme Parp1 (poly(ADP)-ribose polymerase) and its product poly(ADP)-ribose(PAR) accumulate at sites of DNA damage as part of a repair process [9]. This, in turn,directs recruitment and activation of the chromatin remodeling protein ALC1 that maycause nucleosome remodeling [10]. Nucleosome remodeling has been found to cause, forinstance, epigenetic silencing of DNA repair gene MLH1 [11]. Chemicals previously iden‐tified as DNA damaging agents, including benzene, hydroquinone, styrene, carbon tetra‐chloride and trichloroethylene, were shown to cause considerable hypomethylation ofDNA, some through the activation of oxidative stress pathways [12]. Dietary agents alsohave been shown to affect DNA methylation or histone modification by numerous path‐ways [13]. Recent evidence indicates that epimutations occur in DNA repair genes thatreduce their function. Epimutations in DNA repair genes allow DNA damages to accu‐mulate, and are a cause of progression to cancer [14].

2. DNA damages are frequent, and DNA repair processes can beoverwhelmed

Tens of thousands of DNA damages occur per day per cell, on average, in humans, due toreactive molecules produced by metabolism or by hydrolytic reactions in the warm aqueouscellular media. Some types of such endogenous damages, and their rates of occurrence, areshown in Table 1.

A considerable number of other types of endogenous DNA damages have been identified,many of which are mutagenic. These include propano-, etheno- and malondialdehyde-de‐rived DNA adducts, base propenals, estrogen-DNA adducts, alkylated bases, deaminationof each of cytosine, adenine and guanine (to form uracil, hypoxanthine and xanthine, re‐spectively) and adducts formed with DNA by reactive carbonyl species [15].

While there are repair pathways that act on these DNA damages, the repair processes arenot 100% efficient, and further damages occur even as current DNA damages are being re‐paired. Thus there is a steady state level of many DNA damages, reflecting the efficiencies ofrepair and the frequencies of occurrence. For instance, Helbock et al. [16] estimated thesteady state level of oxidative adducts in rat liver as 24, 000 adducts per cell in young ratsand 66, 000 adducts per cell in old rats. Nakamura and Swenberg [17] determined the num‐ber of AP sites (apurinc and apyrimidinic sites) in normal tissues of the rat (i.e. in lung, kid‐ney, liver, testis, heart, colon and brain). The data indicated that the number of AP sitesranged from about 50, 000 per cell in liver, kidney and lung to about 200, 000 per cell in thebrain. These steady state numbers of AP sites in genomic DNA were considered to representthe balance between formation and repair of AP sites.

New Research Directions in DNA Repair416

Page 5: DNA Damage, DNA Repair and Cancer - InTech - Opencdn.intechopen.com/pdfs/43929/InTech-Dna_damage... · Chapter 16 DNA Damage, DNA Repair and Cancer Carol Bernstein, Anil R. Prasad,

DNA damages Reported rate of occurrence Ref.

Oxidative

86,000 per cell per day in rats [18]

10,000 per cell per day in humans

100,000 per cell per day in rats[19]

11,500 per cell per day for humans

74,000 per cell per day for rats[16]

Specific oxidative damage products 8-

hydroxyguanine, 8-hydroxydeoxyguanosine, 5-

(hydroxymethyl) uracil

2,800 per cell per day in humans

34,800 per cell per day in mice[20]

Depurinations

10,000 per cell during 20-hour generation period [21]

13,920 per cell per day (580/cell/hr) [22]

2,000 to 10,000 per cell per day [23,24]

9,000 per cell per day [25]

Depyrimidinations

500 pyrimidines per cell during 20-hour generation

period[21]

696 per cell per day (29/cell/hr) [22]

Single-strand breaks 55,200 per cell per day (2,300/cell/hr) [22]

Double-strand breaks

~10 per cell cycle in humans [26]

~50 per cell cycle in humans [27]

O6-methylguanine 3,120 per cell per day (130/cell/hr) [22]

Cytosine deamination 192 per cell per day (8/cell/hr) [22]

Table 1. DNA damages due to natural endogenous causes in mammalian cells

DNA Damage, DNA Repair and Cancerhttp://dx.doi.org/10.5772/53919

417

Page 6: DNA Damage, DNA Repair and Cancer - InTech - Opencdn.intechopen.com/pdfs/43929/InTech-Dna_damage... · Chapter 16 DNA Damage, DNA Repair and Cancer Carol Bernstein, Anil R. Prasad,

DNA repair pathways are usually able to keep up with the endogenous damages in rep‐licating cells, in part by halting DNA replication at the site of damage until repair canoccur [28, 29]. In contrast, non-replicating cells have a build-up of DNA damages, caus‐ing aging [30, 31].

However, some exogenous DNA damaging agents, such as those in tobacco smoke, dis‐cussed below, may overload the repair pathways, either with higher levels of the same typeof DNA damages as those occurring endogenously or with novel types of damage that arerepaired more slowly. In addition, if DNA repair pathways are deficient, due to inheritedmutations or sporadic somatic epimutations in DNA repair genes in replicating somaticcells, unrepaired endogenous and exogenous damages will increase due to insufficient re‐pair. Increased DNA damages would likely give rise to increased errors of replication pastthe damages (by trans-lesion synthesis) or increased error prone repair (e.g. by non-homolo‐gous end-joining), causing mutations. Increased mutations that activate oncogenes, inacti‐vate tumor suppressor genes, cause genomic instability or give rise to other drivermutations in replicating cells would increase the risk of cancer.

3. Cancers are often caused by exogenous DNA damaging agents

Cancer incidence, in different areas of the world, varies considerably. Thus, the incidence ofcolon cancer among Black Native-Africans is less than 1 person out of 100, 000, while amongmale Black African-Americans it is 72.9 per 100, 000, and this difference is likely due to dif‐ferences in diet [32, 33]. Rates of colon cancer incidence among populations migrating fromlower-incidence to higher-incidence countries change rapidly, and within one generationcan reach the rate in the higher-incidence country. This is observed, for instance, in migrantsfrom Japan to Hawaii [34].

The most common cancers for men and women and their rates of incidence per 100, 000,averaged over the more developed areas and less developed areas of the world, are shownin Table 2 (from [35]). Overall, worldwide, cancer incidence in all organs combined is 300.1per 100, 000 per year in more developed areas and 160.3 per 100, 000 per year in less devel‐oped areas [35]. The differences in cancer incidence between more developed areas of theworld and less developed areas are likely due, in large part, to differences in exposure toexogenous carcinogenic factors. The lowest rates of cancers in a given organ (Table 2) maybe due, at least in part, to endogenous DNA damages (as described in the previous section)that cause errors of replication (trans-lesion synthesis) or error prone repair (e.g. non-homol‐ogous end-joining), leading to carcinogenic mutations. The higher rates (Table 2) are likelylargely attributable to exogenous factors, such as higher rates of tobacco use or diets higherin saturated fats that directly, or indirectly, increase the incidence of DNA damage.

It is interesting to note in Table 2 that, in cases where cancers occur in the same organs ofmen and women, men consistently have a higher rate of cancer than women. The basis forthis is currently unknown.

New Research Directions in DNA Repair418

Page 7: DNA Damage, DNA Repair and Cancer - InTech - Opencdn.intechopen.com/pdfs/43929/InTech-Dna_damage... · Chapter 16 DNA Damage, DNA Repair and Cancer Carol Bernstein, Anil R. Prasad,

More developed areas Less developed areas

Incidence Mortality Incidence Mortality

Breast (women) 66.4 15.3 27.3 10.8

Prostate (men) 62.0 10.6 12.0 5.6

Lung (men) 47.4 39.4 27.8 24.6

Lung (women) 18.6 13.6 11.1 9.7

Colorectum (men) 37.6 15.1 12.1 6.9

Colorectum (women) 24.2 9.7 9.4 5.4

Esophagus (men) 6.5 5.3 11.8 10.1

Esophagus (women) 1.2 1.0 5.7 4.7

Stomach (men) 16.7 10.4 21.1 16.0

Stomach (women) 7.3 4.7 10.0 8.1

Liver (men) 8.1 7.2 18.9 17.4

Liver (women) 2.7 2.5 7.6 7.2

Bladder (men) 16.6 4.6 5.4 2.6

Bladder (women) 3.6 1.0 1.4 0.7

Cervix/Uterine (women) 12.9 2.4 5.9 1.7

Kidney (men) 11.8 4.1 2.5 1.3

Kidney (women) 5.8 1.7 1.4 0.8

Non-Hodgkin lymphoma (men) 10.3 3.6 4.2 3.0

Non-Hodgkin lymphoma

(women)7.0 2.2 2.8 1.9

Melanoma (men) 9.5 1.8 0.7 0.3

Melanoma (women) 8.6 1.1 0.6 0.3

Ovarian (women) 9.4 5.1 5.0 3.1

Table 2. Incidence and mortality rates for the most common cancers in age standardized rates per 100, 000 (excludingnon-melanoma skin cancer) (Adapted from Jemal et al. [35]).

4. Exogenous DNA damaging agents in carcinogenesis

Carcinogenic exogenous factors have been identified as a major cause of many common can‐cers, including cancers of the lung, colorectum, esophagus, stomach, liver, cervix/uterus andmelanoma. Often such exogenous factors have been shown to cause DNA damage, as de‐scribed below.

DNA Damage, DNA Repair and Cancerhttp://dx.doi.org/10.5772/53919

419

Page 8: DNA Damage, DNA Repair and Cancer - InTech - Opencdn.intechopen.com/pdfs/43929/InTech-Dna_damage... · Chapter 16 DNA Damage, DNA Repair and Cancer Carol Bernstein, Anil R. Prasad,

5. Exogenous DNA damaging agents in lung cancer

In both developed and undeveloped countries, lung cancer is the most frequent causeof cancer mortality (Table 2, data for men and women combined). Lung cancer is large‐ly caused by tobacco smoke, since risk estimates for lung cancer indicate that, in theUnited States, tobacco smoke is responsible for 90% of lung cancers. Also implicated inlung cancer (and somewhat overlapping with smoking) are occupational exposure tocarcinogens (approximately 9 to 15%), radon (10%) and outdoor air pollution (perhaps 1to 2%) [36].

Acrolein 122.4

Formaldehyde 60.5

Acrylonitrile 29.3

1,3-butadiene 105.0

Acetaldehyde 1448.0

Ethylene oxide 7.0

Isoprene 952.0

Benzo[a]pyrene 0.014

Table 3. Weight, in μg per cigarette, of several likely carcinogenic DNA damaging agents in tobacco smoke (from [37]Cunningham et al., 2011])

Tobacco smoke is a complex mixture of over 5, 300 identified chemicals, of which 150 areknown to have specific toxicological properties (see partial summary by Cunningham [37]).A “Margin of Exposure” approach has recently been established to determine the most im‐portant exogenous carcinogenic factors in tobacco smoke [37]. This quantitative-type ofmeasurement is based on published dose response data for mutagenicity or carcinogenicityand the concentrations of these components in tobacco smoke (Table 3). Using the “Marginof Exposure” approach, Cunningham et al. [37] found the most important tumorigenic com‐pounds in tobacco smoke to be, in order of importance, acrolein, formaldehyde, acryloni‐trile, 1, 3-butadiene, acetaldehyde, ethylene oxide and isoprene.

Acrolein, the first agent in Table 3, is the structurally simplest α, β-unsaturated aldehyde(Figure 2). It can rapidly penetrate through the cell membrane and bind to the nucleophilicN2-amine of deoxyguanine (dG) followed by cyclization of N1, to give the exocyclic DNAadduct α-hydroxy-1, N2-propano-2’-deoxyguanine (α-HOPdG) (shown in Figure 2) and an‐other product designated γ-HOPdG. The adducts formed by acrolein are a major type ofDNA damage caused by tobacco smoke, and acrolein has been found to be mutagenic [38].

In tobacco smoke, acrolein has a concentration >8, 000 fold higher than benzo[a]pyrene (re‐viewed in [38]), with 122.4 μg of acrolein per cigarette. Benzo[a]pyrene has long beenthought to be an important carcinogen in tobacco smoke [39]. As reviewed by Alexandrov et

New Research Directions in DNA Repair420

Page 9: DNA Damage, DNA Repair and Cancer - InTech - Opencdn.intechopen.com/pdfs/43929/InTech-Dna_damage... · Chapter 16 DNA Damage, DNA Repair and Cancer Carol Bernstein, Anil R. Prasad,

al. [39], benzo[a]pyrene damages DNA by forming DNA adducts at the N2 position of gua‐nine (similar to where acrolein forms adducts). However, by the “Margin of Exposure” ap‐proach, based on published dose response data and its concentration in cigarette smoke of0.014 μg per cigarette, benzo[a]pyrene is thought to be a much less important mutagen forlung tissue than acrolein and the other six highly likely carcinogens in tobacco smoke listedin Table 3 [37].

The other agents in Table 3 cause DNA damages in different ways. Formaldehyde, the sec‐ond agent in Table 3, primarily causes DNA damage by introducing DNA-protein cross-links. These cross-links, in turn, cause mutagenic deletions or other small-scalechromosomal rearrangements [40] and may also cause mutations through single-nucleotideinsertions [41]. Acrylonitrile, the third agent in Table 3, appears to cause DNA damage indi‐rectly by increasing oxidative stress, leading to increased levels of 8’-hydroxyl-2-deoxygua‐nosine (8-OHdG) in DNA [42]. Oxidative stress also causes lipid peroxidation that generatesmalondialdehyde (MDA), and MDA forms DNA adducts with guanine, adenine and cyto‐sine [43]. The fourth agent in Table 3, 1, 3-butadiene, causes genotoxicity both directly byforming a DNA adduct as well as indirectly by causing global loss of DNA methylation andhistone methylation leading to epigenetic alterations [44]. The fifth agent in Table 3, acetal‐dehyde, reacts with 2’-deoxyguanosine in DNA to form DNA adducts [45]. The sixth agentin Table 3, ethylene oxide, forms mutagenic hydroxyethyl DNA adducts with adenine andguanine [46]. The seventh agent in Table 3, isoprene, is normally produced endogenously byhumans, and is the main hydrocarbon of non-smoking human breath [47]. However, smok‐ing one cigarette causes an increase of breath isoprene levels by an average of 70% [48]. Iso‐prene, after being metabolized to mono-epoxides, causes DNA damage measured as singleand double strand breaks in DNA [49].

A large number of studies have been published in which the levels and characteristics ofDNA adducts in the lung and bronchus of smokers and non-smokers have been compared,as reviewed by Phillips [50]. In most of these studies, significantly elevated levels of DNAadducts were detected in the peripheral lung, bronchial epithelium or bronchioalveolar lav‐age cells of the smokers, especially for total bulky DNA adducts. As further discussed byPhillips [50], mean levels of DNA adducts in ex-smokers (usually with at least a 1 year inter‐val since smoking cessation) are found generally to be intermediate between the levels ofsmokers and life-long non-smokers. From these comparisons, the half-life of some DNA ad‐ducts in lung tissue are estimated to be ~1–2 years.

6. Exogenous DNA damaging agents in colorectal cancer

Up to 20% of current colorectal cancers in the United States may be due to tobacco smoke[51]. Presumably tobacco smoke causes colon cancer due to the DNA damaging agents de‐scribed above for lung cancer. These agents may be taken up in the blood and carried to or‐gans of the body.

DNA Damage, DNA Repair and Cancerhttp://dx.doi.org/10.5772/53919

421

Page 10: DNA Damage, DNA Repair and Cancer - InTech - Opencdn.intechopen.com/pdfs/43929/InTech-Dna_damage... · Chapter 16 DNA Damage, DNA Repair and Cancer Carol Bernstein, Anil R. Prasad,

Figure 2. Reaction of acrolein with deoxyguanosine

The human colon is exposed to many compounds that are either of direct dietary origin or re‐sult from digestive and/or microbial processes. Four different classes of colonic mutageniccompounds were analysed by de Kok and van Maanen [52] and evaluated for fecal mutagenici‐ty. These included (1) pyrolysis compounds from food (heterocyclic aromatic amines and poly‐cyclic aromatic hydrocarbons), (2) N-nitroso-compounds (from high meat diets, from drinkingwater with high nitrates or produced during ulcerative colitis), (3) fecapentaenes (produced bythe colonic bacteria Bacteriodes in the presence of bile acids) and (4) bile acids (increased in thecolon in response to a high fat diet and metabolized to genotoxic form by bacteria in the colon).Many of these diet-related mutagenic compounds were analysed by Pearson et al. [53] in termsof their presence in fecal water, and their effect on the cytotoxic or genotoxic activity of fecalwater. Evidence in both of these studies was insufficient to evaluate the colorectal cancer risk asa result of specific exposures in quantitative terms.

However, substantial evidence implicates bile acids (the 4th possibility above) in colon caner.Bernstein et al. [54], summarized 12 studies indicating that the bile acids deoxycholic acid(DCA) and/or lithocholic acid (LCA) induce production of DNA damaging reactive oxygenspecies and/or reactive nitrogen species in colon cells of animal or human origin. They alsotabulated 14 studies showing that DCA and LCA induce DNA damage in colon cells. In ad‐dition to causing DNA damage, bile acids may also generate genomic instability by causing

New Research Directions in DNA Repair422

Page 11: DNA Damage, DNA Repair and Cancer - InTech - Opencdn.intechopen.com/pdfs/43929/InTech-Dna_damage... · Chapter 16 DNA Damage, DNA Repair and Cancer Carol Bernstein, Anil R. Prasad,

mitotic perturbations and reduced expression of spindle checkpoint proteins, giving rise tomicro-nuclei, chromosome bridges and other structures that are precursors to aneuploidy[55]. Furthermore, at high physiological concentrations, bile acids cause frequent apoptosis,and those cells in the exposed populations with reduced apoptosis capability tend to surviveand selectively proliferate [54, 56]. Cells with reduced ability to undergo apoptosis in re‐sponse to DNA damage would tend to accumulate mutations when replication occurs pastthose damages, and such cells may give rise to colon cancers. In addition, 7 epidemiologicalstudies between 1971 and 1990 (reviewed by Bernstein et al. [54]), found that fecal bile acidconcentrations are increased in populations with a high incidence of colorectal cancer. Asimilar 2012 epidemiological study showed that concentrations of fecal LCA and DCA, re‐spectively, were 4-fold and 5-fold higher in a population at 65-fold higher risk of colon can‐cer compared to a population at lower risk of colon cancer [32]. This evidence points to bileacids DCA and LCA as centrally important DNA-damaging carcinogens in colon cancer.

Dietary total fat intake and dietary saturated fat intake is significantly related to incidence ofcolon cancer [57]. Increasing total fat or saturated fat in human diets results in increases inDCA and LCA in the feces [58, 59], indicating increased contact of the colonic epitheliumwith DCA and LCA. Bernstein et al. [60] added the bile acid DCA to the standard diet ofwild-type mice. This supplement raised the level of DCA in the feces of mice from the stand‐ard-diet fed mouse level of 0.3 mg DCA/g dry weight to 4.6 mg DCA/g dry weight, a levelsimilar to that for humans on a high fat diet of 6.4 mg DCA/g dry weight. After 8 or 10months on the DCA-supplemented diet, 56% of the mice developed invasive colon cancer.This directly indicates that DCA, a DNA damaging agent, at levels present in humans after ahigh fat diet, can cause colorectal cancer.

7. Exogenous DNA damaging agents implicated in other major cancers

It is beyond the scope of this chapter to detail the evidence implicating DNA damagingagents as etiologic agents in all of the significant cancers. Therefore, in Table 4 we indicatewith a single reference the major DNA damaging agent in five additional prevalent cancers,in order to illustrate the generality of exogenous DNA damaging agents as causes of cancer.In particular, we point out, as reviewed by Handa et al. [61], Helicobacter pylori infection in‐creases the production of reactive oxygen and reactive nitrogen species (RNS) in the humanstomach, which, in turn, significantly increases DNA damage in the gastric epithelial cells.Thus, H. pylori infection acts as a DNA damaging agent. In the case of human papillomavi‐rus (HPV) infection, Wei et al. [62] showed that cervical cells could resist RNS stress whennot infected with HPV. However, cervical cells infected by HPV and exposed to RNS hadhigher levels of DNA double strand breaks as well as a higher mutation rate. This appearedto occur due to the ability of HPV to greatly reduce protein expression of the DNA damagerepair/response gene P53 when infected cells were stressed by RNS. Since reduced P53 ex‐pression leads to greater RNS-induced DNA damage, HPV infection acts as a DNA damag‐ing agent in the presence of RNS stress.

DNA Damage, DNA Repair and Cancerhttp://dx.doi.org/10.5772/53919

423

Page 12: DNA Damage, DNA Repair and Cancer - InTech - Opencdn.intechopen.com/pdfs/43929/InTech-Dna_damage... · Chapter 16 DNA Damage, DNA Repair and Cancer Carol Bernstein, Anil R. Prasad,

Cancer Exogenous DNA damaging agent Ref.

Esophagus Bile acids [63]

Stomach Helicobacter pylori infection [61]

Liver Aspergillus metabolite aflatoxin B(1) [64]

Cervix/UterusHuman papillomavirus plus increased nitric oxide from tobacco smoke or

other infection[62]

Melanoma UV light from solar radiation [65]

Table 4. Selected cancers and relevant implicated exogenous DNA damaging agents

8. Deficient DNA repair due to a germ line mutation allows DNAdamages to increase, leading to increased frequencies of mutation,epimutation and cancer

Expression of DNA repair genes may be reduced by inherited germ line mutations or genet‐ic polymorphisms, or by epigenetic alterations or mutations in somatic cells, and these re‐ductions may substantially increase the risk of cancer. Overall, about 30% of cancers areconsidered to be familial (largely due to inherited germ line mutations or genetic polymor‐phisms) and 70% are considered to be sporadic [66].

In 2 overlapping databases [67, 68] 167 and169 human genes (depending on the database)are listed that are directly employed in DNA repair or influence DNA repair processes. Thelists were originally devised by Wood et al. [69, 70]. The genes are distributed in groups ofDNA repair pathways and in related functions that affect DNA repair (Table 5). Bernstein etal. [71] illustrate many of the steps and order of action of the gene products involved for thefirst five DNA repair pathways listed in Table 5.

Individuals with an inherited impairment in DNA repair capability are often at considerablyincreased risk of cancer. If an individual has a germ line mutation in a DNA repair gene or aDNA damage response gene (that recognizes DNA damage and activates DNA repair), usu‐ally one abnormal copy of the gene is inherited from one of the parents and then the othercopy is inactivated at some later point in life in a somatic cell. The inactivation may be due,for example, to point mutation, deletion, gene conversion, epigenetic silencing or othermechanisms [72]. The protein encoded by the gene will either not be expressed or be ex‐pressed in a mutated form. Consequently the DNA repair or DNA damage response func‐tion will be deficient or impaired, and damages will accumulate. Such DNA damages cancause errors during DNA replication or inaccurate repair, leading to mutations that can giverise to cancer.

Increased oxidative DNA damages also cause increased gene silencing by CpG island hy‐permethylation, a form of epimutation. These oxidative DNA damages induce formationand relocalization of a silencing complex that may result in cancer-specific aberrant DNA

New Research Directions in DNA Repair424

Page 13: DNA Damage, DNA Repair and Cancer - InTech - Opencdn.intechopen.com/pdfs/43929/InTech-Dna_damage... · Chapter 16 DNA Damage, DNA Repair and Cancer Carol Bernstein, Anil R. Prasad,

methylation and transcriptional silencing [73]. As pointed out above, the enzyme Parp1(poly(ADP)-ribose polymerase) and its product poly(ADP)-ribose (PAR) accumulate at sitesof DNA damage as part of a repair process [9], recruiting chromatin remodeling proteinALC1, causing nucleosome remodeling [10] that has been shown to direct epigenetic silenc‐ing of DNA repair gene MLH1 [11]. If silencing of genes necessary for DNA repair occurs,the repair of further DNA damages will be deficient and more damages will accumulate.Such additional DNA damages will cause increased errors during DNA synthesis, leadingto mutations that can give rise to cancer.

Number of genes listed in the two

databases

Homologous Recombinational Repair (HRR) 21,21

Non-homologous End Joining (NHEJ) 8,7

Nucleotide Excision Repair (NER) 30,29

Base Excision Repair (including PARP enzymes) (BER) 19,20

Mis-Match Repair (MMR) 11,10

Fanconi Anemia (FANC) [affects HRR (above) and translesion synthesis (TLS)] 10,16

Direct reversal of damage 3,3

DNA polymerases (act in various pathways) 17,15

Editing and processing nucleases (act in various pathways) 6,8

Ubiquitination and modification/Rad6 pathway including TLS 11,5

DNA damage response 12,14

Modulation of nucleotide pools 3,3

Chromatin structure 2,3

Defective in diseases and syndromes 4,5

DNA-topoisomerase crosslinks 2,1

Other genes 8,9

Table 5. DNA repair pathways and other processes affecting DNA repair [67, 68]

DNA Damage, DNA Repair and Cancerhttp://dx.doi.org/10.5772/53919

425

Page 14: DNA Damage, DNA Repair and Cancer - InTech - Opencdn.intechopen.com/pdfs/43929/InTech-Dna_damage... · Chapter 16 DNA Damage, DNA Repair and Cancer Carol Bernstein, Anil R. Prasad,

9. Inherited mutations in genes employed in DNA repair that give rise tosyndromes characterized by increased risk of cancer.

Table 6 lists 36 genes for which an inherited mutation results in an increased risk of cancer.The proteins encoded by 35 of these genes are involved in DNA repair and in some casesalso in other aspects of the DNA damage response such as cell cycle arrest and apoptosis.The polymerase coded for by the 36th gene, XPV (POLH), is involved in bypass (rather thanrepair) of DNA damage, called translesion synthesis. The genes listed in Table 6, when mu‐tated in the germ line, give rise to a considerably increased lifetime risk of cancer, of up to100% (e.g. p53 mutations [74]). Thus defects in DNA repair cause progression to cancer.

In addition to mutations in genes that may substantially raise lifetime cancer risk, there appear tobe many weakly effective genetically inherited polymorphisms [single nucleotide polymo‐phisms (SNPs) and copy number variants (CNVs)]. By the HapMap Project, more than 3 millionSNPs have been found, and by Genome Wide Association studies (GWAs), about 30 SNPs werefound to increase risk of cancers. However the added risk of cancer by these SNPs is usuallysmall, i.e. less than a factor of 2 increase [75]. A large twin study [66], involving 44, 788 pairs oftwins, evaluated the risk of the same cancer before the age of 75 for monozygotic twins (identicalgenomes with the same polymorphisms) and dizygotic twins (having a 50% chance of the samepolymorphisms). If one twin had colorectal, breast or prostate cancer, the monozygotic twin hadan 11 to 18 percent chance of developing the same cancer while the dizygotic twin had only a 3 to9% risk. The differences in monozygotic and dizygotic rates of paired cancer were not significantfor the other 24 types of cancer evaluated in this study. Polymorphisms of the DNA repair geneERCC1 will be discussed below in relation to targeted chemotherapy.

10. Epimutations may repress DNA repair gene expression, allowingDNA damages to increase, leading to increased frequency of furtherepimutation, mutation and cancer

While germ line (familial) mutations in DNA repair genes cause a high risk of cancer, somat‐ic mutations in DNA repair genes are rarely found in sporadic (non-familial) cancers [4].Much more often, DNA repair genes are found to have epigenetic alterations in cancers.

One example of the epigenetic down-regulation of a DNA repair gene in cancers comes fromstudies of the MMR protein MLH1. Truninger et al. [76] assessed 1, 048 unselected consecutivecolon cancers. Of these, 103 were deficient in protein expression of MLH1, with 68 of these can‐cers being sporadic (the remaining MLH1 deficient cancers were due to germ line mutations). Ofthe 68 sporadic MLH1 protein-deficient colon cancers, 65 (96%) were found to be deficient due toepigenetic methylation of the CpG island of the MLH1 gene. Deficient protein expression ofMLH1 may also have been caused, in the remaining 3 sporadic MLH1 protein-deficient cancers(which did not have germ line mutations), by over expression of the microRNA miR-155. WhenmiR-155 was transfected into cells it caused reduced expression of MLH1 [77]. Overexpressionof miR-155 was found in colon cancers in which protein expression of MLH1 was deficient andthe MLH1 gene was neither mutated nor hypermethylated in its CpG island [77].

New Research Directions in DNA Repair426

Page 15: DNA Damage, DNA Repair and Cancer - InTech - Opencdn.intechopen.com/pdfs/43929/InTech-Dna_damage... · Chapter 16 DNA Damage, DNA Repair and Cancer Carol Bernstein, Anil R. Prasad,

DNA repair gene(s)Encoded

protein

Repair pathway(s)

affectedRef.

Cancers with

increased riskRef.

breast cancer 1 & 2BRCA1,

BRCA2

HRR of double strand breaks

and daughter strand gaps[85] Breast, Ovarian [86]

ataxia telangiectasia mutated ATM

Different mutations in ATM

reduce HRR, single strand

annealing (SSA), NHEJ or

homology directed double

strand break rejoining

(HDR)

[87]Leukemia, Lymphoma,

Breast[87,88]

Nijmegen breakage syndrome NBS NHEJ [89] Lymphoid cancers [89]

meiotic recombination 11 MRE11 HRR and NHEJ [90] Breast [91]

Bloom’s Syndrome (helicase) BLM HRR [92]

Leukemia, Lymphoma,

Colon, Breast, Skin,

Auditory canal, Tongue,

Esophagus, Stomach,

Tonsil, Larynx, Lung,

Uterus

[93]

Werner Syndrome (helicase) WRN HRR, NHEJ, long patch BER [94]

Soft tissue sarcoma,

Colorectal, Skin,

Thyroid,

Pancreatic

[95]

Rothman Thomson syndrome

Rapadilino syndrome

Baller Gerold syndrome

RECQ4 Helicase likely active in HRR [96]

Basal cell carcinoma,

Squamous cell

carcinoma,

Intraepidemial

carcinoma

[97]

Fanconi’s anemia gene FANC

A,B,C,D1,D2,E,F,G,I,J,L,M,NFANCA etc. HRR and TLS [98]

Leukemia, Liver tumors,

Solid tumors many

areas

[99]

xeroderma pigmentosa

C, E [DNA damage binding

protein 2 (DDB2)]

XPC

XPE

Global genomic NER repairs

damage in both transcribed

and untranscribed DNA

[100,

101]

Skin cancer (melanoma

and non-melanoma)

[100,

101]

xeroderma pigmentosa

A, B, D, F, G

XPA XPB

XPD XPF

XPG

Transcription coupled NER

repairs the transcribed

strands of transcriptionally

active genes

[102]

Skin cancer (melanoma

and non-melanoma),

Central nervous system

cancers

[102]

xeroderma pigmentosa V (also

called polymerase H)

XPV

(POLH)Translesion Synthesis (TLS) [102]

Skin cancer (melanoma

and non-melanoma)[102]

DNA Damage, DNA Repair and Cancerhttp://dx.doi.org/10.5772/53919

427

Page 16: DNA Damage, DNA Repair and Cancer - InTech - Opencdn.intechopen.com/pdfs/43929/InTech-Dna_damage... · Chapter 16 DNA Damage, DNA Repair and Cancer Carol Bernstein, Anil R. Prasad,

DNA repair gene(s)Encoded

protein

Repair pathway(s)

affectedRef.

Cancers with

increased riskRef.

mutS (E. coli) homolog 2

mutS (E. coli) homolog 6

mutL (E. coli) homolog 1

postmeiotic segregation

increased 2 (S. cerevisiae)

MSH2

MSH6

MLH1

Pms2

MMR [76]Colorectal, endometrial.

ovarian[103]

mutY homolog (E. coli) MUTYHBER of A mispaired with

8OHdG, G, FapydG and C[104] Colon [105]

ataxia telaniectsia and RAD3

relatedATR

DNA damage response

likely affects HRR, not NHEJ[106] Oropharyngeal cancer [107]

Li Fraumeni syndrome P53

HRR, BER, NER and DNA

Damage Response for those

and for NHEJ and MMR

[108]

Sarcoma, Breast, Lung,

Skin, Pancreas,

Leukemia, Brain

[74]

Table 6. Inherited mutations in DNA repair genes that increase the risk of cancer

Another example of the epigenetic down-regulation of a DNA repair gene in cancer comes fromstudies of the direct reversal of methylated guanine bases by methyl guanine methyl transferase(MGMT). In the most common form of brain cancer, glioblastoma, the DNA repair gene MGMTis epigenetically methylated in 29% [78] to 66% [79] of tumors, thereby reducing protein expres‐sion of MGMT. However, for 28% of glioblastomas, the MGMT protein is deficient but theMGMT promoter is not methylated [79]. Zhang et al. [78] found, in the glioblastomas withoutmethylated MGMT promoters, that the level of microRNA miR-181d is inversely correlatedwith protein expression of MGMT and that the direct target of miR-181d is the MGMT mRNA3’UTR (the three prime untranslated region of MGMT mRNA), though they indicated that othermiRNAs may also be involved in the reduction of protein expression of MGMT.

Almost all DNA repair deficiencies found, so far, in sporadic cancers, and in precanceroustissues surrounding cancers (field defects) are due to epigenetic changes. Examples of suchepigenetic alterations in DNA repair genes in different types of cancer are shown in Table 7.A recent review [80] lists 41 reports (mostly not overlapping with those listed in Table 7)indicating methylation of 20 DNA repair genes in various cancers. In Table 7 data are alsoshown on DNA repair gene deficiencies for the field defects associated with colorectal, gas‐tric, laryngeal and non-small cell lung cancer.

As summarized above, epimutations can result from oxidative DNA damages. Such damagescause formation and relocalization of a silencing complex that in turn causes increased gene si‐lencing by CpG island hypermethylation [73]. Epigenetic nucleosome remodeling during DNArepair can also silence gene expression [11]. When CpG island methylation or nucleosome re‐modeling or other types of epigenetic alterations (e.g. micro RNAs or histone modifications) in‐hibit DNA repair genes, more damages will accumulate. Accumulated DNA damages causeincreased errors during DNA synthesis and repair. Thus epigenetic deficiencies in DNA repairgenes can have a cascading effect (a mutator phenotype), leading to genomic instability and ac‐cumulation of mutations and epimutations that can give rise to cancer.

New Research Directions in DNA Repair428

Page 17: DNA Damage, DNA Repair and Cancer - InTech - Opencdn.intechopen.com/pdfs/43929/InTech-Dna_damage... · Chapter 16 DNA Damage, DNA Repair and Cancer Carol Bernstein, Anil R. Prasad,

Cancer

Epigenetic changes

in cancer

(mechanism)

% sporadic cancers

with epimutations

Epigenetic

changes in field

defect

(mechanism)

% field defects

with epi-

mutations

Ref.

BreastBRCA1 (CGI*)

13% unselected

[108]67% medullary

55% mucinous

WRN (CGI) 17% unselected [2]

Ovarian BRCA1 (CGI)

31% of those with

loss of

heterozygosity

[108]

Colorectal

WRN (CGI) 38% [2]

MGMT (CGI) 46% MGMT (CGI) 23% [109]

MGMT (CGI) 90%[110]

MLH1 (CGI) 65%

MLH1 (CGI) 10% [76]

MLH1 (CGI) 2%

[111]MSH2 (CGI) 13% MSH2 5%

MGMT (CGI) 47% MGMT 11%

ERCC1 100% ERCC1 40%

[112]PMS2 88% PMS2 50%

XPF 55% XPF 40%

GastricMGMT (CGI) 88% MGMT (CGI) 29% [113]

WRN (CGI) 25% [2]

Esophageal

squamous cell

carcininoma

MLH1 (CGI) 49%

[114,115]MLH2 (CGI) 35%

MGMT (CGI) 41%

Larynx MGMT (CGI) 54% MGMT (CGI) 38% [116]

DNA Damage, DNA Repair and Cancerhttp://dx.doi.org/10.5772/53919

429

Page 18: DNA Damage, DNA Repair and Cancer - InTech - Opencdn.intechopen.com/pdfs/43929/InTech-Dna_damage... · Chapter 16 DNA Damage, DNA Repair and Cancer Carol Bernstein, Anil R. Prasad,

Cancer

Epigenetic changes

in cancer

(mechanism)

% sporadic cancers

with epimutations

Epigenetic

changes in field

defect

(mechanism)

% field defects

with epi-

mutations

Ref.

Non-small cell LungWRN (CGI) 38% [2]

MGMT (CGI) 70% MGMT (CGI) 40% [117]

Prostate WRN (CGI) 20% [2]

Thyroid WRN (CGI) 13% [2]

Non-Hodgkin

lymphomaWRN (CGI) 24% [2]

Leukemias WRN (CGI) 5-10% [2]

Chondrosarcomas WRN (CGI) 33% [2]

Osteosarcomas WRN (CGI) 11% [2]

Brain glioblastomaMGMT (CGI) 51% [118]

MGMT (miRNA) 28% [78]

Liver hepatocellular

carcinoma

P53 (non-CGI

promoter site specific

methylation)

100% [119]

Papillary thyroid

(tested 23 DNA repair

genes for CGI)

MLH1 (CGI) 21%

[120]PCNA (CGI) 13%

OGG1 (CGI) 2%

*CGI=CpG island methlyation

Table 7. Examples of epigenetic alterations (epimutations) of DNA repair genes in cancers and in field defects, withmechanisms indicated where known.

Deficiencies in DNA repair genes cause increased mutation rates. Mutations rates increasein MMR defective cells [81, 82] and in HRR defective cells [83]. Chromosomal rearrange‐ments and aneuploidy also increase in HRR defective cells [84]. Thus, deficiency in DNA re‐pair causes genomic instability and genomic instability is the likely main underlying causeof the genetic alterations leading to tumorigenesis. Deficient DNA repair permits the acquis‐ition of a sufficient number of alterations in tumor suppressor genes and oncogenes to fuelcarcinogenesis. Deficiencies in DNA repair appear to be central to the genomic and epige‐nomic instability characteristic of cancer.

New Research Directions in DNA Repair430

Page 19: DNA Damage, DNA Repair and Cancer - InTech - Opencdn.intechopen.com/pdfs/43929/InTech-Dna_damage... · Chapter 16 DNA Damage, DNA Repair and Cancer Carol Bernstein, Anil R. Prasad,

Figure 3 illustrates the chain of consequences of exposure of cells to endogenous andexogenous DNA damaging agents that lead to cancer. The role of germ line defects inDNA repair genes in familial cancer are also indicated. The large role of DNA damageand consequent epigenetic DNA repair defects leading to sporadic cancer are empha‐sized. The roles of germ line mutation and directly induced somatic mutation in spora‐dic cancer are indicated as well.

Figure 3. The roles of DNA damage, epigenetic deficiencies in DNA repair and mutation in progression to cancer.

DNA Damage, DNA Repair and Cancerhttp://dx.doi.org/10.5772/53919

431

Page 20: DNA Damage, DNA Repair and Cancer - InTech - Opencdn.intechopen.com/pdfs/43929/InTech-Dna_damage... · Chapter 16 DNA Damage, DNA Repair and Cancer Carol Bernstein, Anil R. Prasad,

11. Epigenetic alterations caused by micro RNAs

MicroRNAs (miRNAs) are endogenous non-coding RNAs, 19-25 nucleotides in length, thatcan have substantial effects on DNA repair. miRNAs can either directly or indirectly reduceexpression of DNA repair or DNA damage response genes. As discussed above, over-ex‐pression of miR-155 causes reduced expression of DNA repair protein MLH1, and miR-155is overexpressed in colon cancers [77] (curved arrow in Figure 3). Similarly, miR-181d isoverexpressed in glioblastomas, causing reduced expression of DNA repair protein MGMT[78]. Although miRNAs can epigenetically regulate DNA repair gene expression, the expres‐sion levels of many miRNAs may themselves be subject to epigenetic regulation. One mech‐anism of epigenetic regulation of miRNA expression is hypomethylation of the promoterregion of the DNA sequence that codes for the miRNA. Schnekenburger and Diederich [7]list miR-155 as one of a long list of mi-RNAs whose expression is increased by hypomythy‐lation in colorectal cancers. In particular, hypomethylated miR-155 (the hypomethylationmaking it more active) targets genes MLH1, MSH2 and MSH6, causing each of them to havereduced expression [7].

Wan et al. [121] referred to 6 further DNA repair genes that are directly targeted by miR‐NAs. ATM, RAD52, RAD23B, MSH2, BRCA1 and P53, are each specifically targeted by oneor two of the 8 miRNAs miR-21, miR-24, miR-125b, miR-182, miR-210, miR-373, miR-421and miR-504, with all but miR-210, miR-421 and miR-504 among those identified by Schne‐kenburger and Diederich [7] as overexpressed through epigenetic hypomethylation. Overex‐pression of any one of these miRNAs leads to reduced expression of its target DNA repairgene. Wan et al. [121] further listed 16 DNA damage response genes targeted by specificmiRNAs. Wan et al. [121] indicated miR-15a, miR-16, miR-17, miR-20a, miR-21, miR-24,miR-29, miR-34a, miR-106a, miR-93, miR-124a, miR-125b, miR-192, miR-195, miR-215,miR-182, miR-373 as among those targeting DNA damage response genes. Of these, all butmiR-124a were identified by Schnekenburger and Diederich [7], (and Malumbres [122] fur‐ther identified miR-34a and miR-124a) as being among miRNAs whose expression is subjectto epigenetic alteration in tumors. Other miRNAs whose expression is subject to epimuta‐tion in colorectal cancers (and their target DNA repair or DNA damage response genes) in‐clude miR-17 (E2F1), miR-34b/c (P53), miR-106a (E2F1), miR-200a and miR-200b (MLH1,MSH2) and miR-675 (Rb) [7].

12. Epigenetic alterations caused by chromosome remodeling and histonemodification

Specific miRNAs can also indirectly (and strongly) reduce protein expression of DNA repairgenes through their role in repression of proteins designated High Mobility Group A1(HMGA1) and HMGA2 (the names come from the proteins’ high electrophoretic mobility onacrylamide gels). HMGA1 and HMGA2 cause chromatin remodeling at specific sites inDNA and reduce expression at those sites. In particular, these proteins appear to control

New Research Directions in DNA Repair432

Page 21: DNA Damage, DNA Repair and Cancer - InTech - Opencdn.intechopen.com/pdfs/43929/InTech-Dna_damage... · Chapter 16 DNA Damage, DNA Repair and Cancer Carol Bernstein, Anil R. Prasad,

DNA repair genes BRCA1 and ERCC1. BRCA1 And ERCC1 proteins have key roles in DNArepair, particularly of double-strand breaks and interstrand crosslinks. HMGA1 and HMGA2genes are usually active in embryogenesis, but normally have very low expression levels inadult tissues. Their expression levels in adult tissues are kept low by the actions of specificmiRNAs. If expression of these miRNAs is reduced, then the repressive HMGA1 andHMGA2 proteins become highly expressed and, in particular, can reduce expression ofBRCA1 or ERCC1 respectively.

As reviewed by Resar [123], all HMG proteins share an acidic carboxyl terminus and asso‐ciate with chromatin. As an example, HMGA1A, in particular, has three AT-hook domainsthat allow it to bind to AT-rich regions and recruit an “enhanceosome” that may displacehistones and cause chromosome remodeling and reduce gene expression. Baldassarre et al.[124] showed that HMGA1B protein binds to the promoter region of BRCA1 and inhibitsBRCA1 promoter activity (indicated in Figure 3 as chromatin remodeling causing reducedBRCA1). In 12 surgically removed human breast carcinomas, there was an inverse correla‐tion between HGMA1 protein and BRCA1 mRNA levels. HGMA1 was almost undetectablein normal breast tissue, highly expressed in the tumor samples, and BRCA1 protein wasstrongly diminished in tumor samples. Baldassarre et al. [124] suggested that while only11% of breast tumors had hypermethylation of the BRCA1 gene, 82% of aggressive breastcancer specimens have low BRCA1 protein, and most of these could be due to chromatin re‐modeling by high levels of HMGA1 protein.

Similarly, HMGA2 binds to an ERCC1 promoter site and represses ERCC1 promoter activity[125]. The miRNAs miR-23a, miR-26a and miR-30a inhibit HMG2A protein expression [126]though it has not been reported whether these miRNAs are under epigenetic control. In Fig‐ure 3, one of two dotted lines is used to indicate possible repression of ERCC1 by epigeneti‐cally induced chromatin remodeling.

Resar [123] and Baldassarre et al. [124] summarized reports indicating that HGMA1 is wide‐ly overexpressed in aggressive malignancies including cancers of the thyroid, head andneck, colon, lung, breast, pancreas, hematopoetic system, cervix, uterine corpus, prostateand central nervous system. Palmieri et al. [127] showed that HGMA1 and HMGA2 are tar‐geted (and thus strongly reduced in expression) by miR-15, miR-16, miR-26a, miR-196a2 andLet-7a. The promoter regions associated with miR-16, miR-196a2 and Let-7a miRNAs are ep‐imutated by hypomethylation [7, 122] while Sampath et al. [128] showed, in addition, thatthe coding regions for miR-15 and miR-16 were epigenetically silenced due to histone deace‐tylase activity. Palmieri et al. [127] further showed that these 5 miRNAs are drastically re‐duced in a panel of 41 pituitary adenomas, accompanied by increases in HMGA1 andHMGA2 specific mRNAs. In a more recent study on pituitary adenomas by D’Angelo et al.[129], reduced expression of 18 miRNAs was found, with 5 of them targeting HMGA1 orHMGA2. In this recent study, among the 18 miRNAs with reduced expression, the reducedexpression of miR-26b, miR-34b, miR-432 and miR-592 was known to be due to epigeneticalteration [7, 122]. Thus, epigenetic miRNA silencing, causing strong expression of HMGA1and HMGA2, occurs in many types of cancer and this may be related to reductions found inexpression of DNA repair genes BRCA1, BRCA2 and ERCC1.

DNA Damage, DNA Repair and Cancerhttp://dx.doi.org/10.5772/53919

433

Page 22: DNA Damage, DNA Repair and Cancer - InTech - Opencdn.intechopen.com/pdfs/43929/InTech-Dna_damage... · Chapter 16 DNA Damage, DNA Repair and Cancer Carol Bernstein, Anil R. Prasad,

Suzuki et al. [130], using genome wide profiling, found 174 primary transcription units formiRNAs, called “pri-miRNAs” (large precursor RNAs which may encode multiple miR‐NAs), of which they identified 37 as potential targets for epigenetic silencing. Of these 37pri-miRNAs, 22 were encoded by DNA sequences with CpG islands (all of which were hy‐permethylated in colorectal cancer cells) while the other pri-miRNAs were subject to regula‐tion by epigenetic “activating marks” without evidence of deregulated methylation.

Activating marks are alterations on histones that cause transcriptional activation of thegenes associated with those altered histones (reviewed by Tchou-Wong et al. [131]). In par‐ticular, the nucleosome, the fundamental subunit of chromatin, is composed of 146 bp ofDNA wrapped around an octamer of four core histone proteins (H3, H4, H2A, and H2B).Posttranslational modifications (i.e., acetylation, methylation, phosphorylation, and ubiqui‐tination) of the N- and C-terminal tails of the four core histones play an important role inregulating chromatin biology. These specific histone modifications, and their combinations,are translated, through protein interactions, into distinct effects on nuclear processes, suchas activation or inhibition of transcription. In eukaryotes, methylation of lysine 4 in histoneH3 (H3K4), which interacts with the promoter region of genes, is linked to transcriptionalactivation. There is a strong positive correlation between trimethylation of H3K4, transcrip‐tion rates, active polymerase II occupancy and histone acetylation. Thus trimethylation ofH3K4 is an activating mark.

In addition to pri-miRNAs being regulated by activating marks, some miRNAs appear to bedirectly regulated by these histone modifications. As summarized by Sampath et al. [128],histone deacetylases catalyze the removal of acetyl groups on specific lysines around genepromoters to trigger demethylation of otherwise methylated lysine 4 on histones(H3K4me2/3) and this causes loss of these activating marks, promoting chromatin compac‐tion, and leading to epigenetic silencing. Sampath et al. [128] showed that such histone de‐acetylase activity mediates the epigenetic silencing of miRNAs miR-15a, miR-16, andmiR-29b. As indicated above, miR-15, miR-16 specifically target HGMA1 and HMGA2. IfmiR-15 and miR-16 lose their activating marks, they have reduced expression, causingHGMA1 and HGMA2 to be transcriptionally activated, thus reducing expression of DNA re‐pair genes BRCA1 and ERCC1.

In Figure 3, histone modification and chromatin remodeling are indicated as epigenticallyaltering the expression of many genes in progression to cancer, and specifically causing re‐duced BRCA1 and possibly (as indicated by one dotted line) reduced expression of ERCC1.In addition, a second dotted line is used to indicate possible repression of ERCC1 by an miR‐NA. Klase et al. [132] showed that a particular virally coded miRNA down regulates ERCC1protein expression at the p-body level (a p-body is a cytoplasmic granule “processing body”that interacts with miRNAs to repress translation or trigger degradation of target mRNAs).A survey of human miRNA homology regions to ERCC1 mRNA indicates at least 21 humancoded miRNAs that could act to decrease ERCC1 mRNA translation (shown in MicrocosmTargets [133]). ERCC1 protein expression, assessed by immunohistochemical staining, is de‐ficient due to an epigenetic mechanism in colon cancers [110], and this could be due to ac‐tion of one or more miRNAs, acting directly on ERCC1 mRNA.

New Research Directions in DNA Repair434

Page 23: DNA Damage, DNA Repair and Cancer - InTech - Opencdn.intechopen.com/pdfs/43929/InTech-Dna_damage... · Chapter 16 DNA Damage, DNA Repair and Cancer Carol Bernstein, Anil R. Prasad,

13. Driver mutations and pathways to cancer progression

Recent research indicates a mechanism by which an early driver mutation may cause subse‐quent epigenetic alterations or mutations in pathways leading to cancer. Wang et al. [134]point out that isocitrate dehydrogenase genes IDH1 and IDH2 are the most frequently mu‐tated metabolic genes in human cancer. A gene frequently mutated in cancer is consideredto be a driver mutation [4] so that mutations in IDH1 and IDH2 would be driver mutations.Wang et al. [134] further point out that IDH1 and IDH2 mutant cells produce an excess met‐abolic intermediate, 2-hydroxyglutarate, which binds to catalytic sites in key enzymes thatare important in altering histone and DNA promoter methylation. Thus, mutations in IDH1and IDH2 generate a DNA CpG island methylator phenotype that causes promoter hyper‐methylation and concomitant silencing of tumor suppressor genes such as the DNA repairgenes MLH1, MGMT and BRCA1. As shown in Figure 3, a driver mutation in IDH1 cancause a feedback loop leading to increased DNA repair deficiency, further mutations andepimutations, and consequent accelerated tumor progression.

A study, involving 51 patients with brain gliomas who had two or more biopsies over time,showed that mutation in the IDH1 gene occurred prior to the occurrence of a p53 mutationor a 1p/19q loss of heterozygosity, indicating that IDH1 mutation is an early driver mutation[135]. Work by Turcan et al. [136] showed that IDH1 mutation alone is sufficient to establishthe brain glioma CpG island methylator phenotype. Carillo et al. [137] showed that when anIDH1 mutation was present in glioblastoma tumors, 64% of these were hypermethylated inthe promoter regions of MGMT.

Other initial driver mutations can cause progression to glioblastoma as well. As pointed outabove, increased levels of miR-181d also cause reduced expression of MGMT protein in glio‐blastoma. Nelson et al. [138] indicate that a single type of miRNA may target hundreds ofdifferent mRNAs, causing alterations in multiple pathways. Patients with a glioblastomathat does not harbor an IDH1 mutation have an overall fairly short survival time, while pa‐tients with both mutated IDH1 and methylated MGMT have a subtype of glioblastoma witha much longer survival time (implying a different pathway of cancer progression) [137].

An IDH1 mutation that gives rise to a CpG island methylator phenotype that causes pro‐moter hypermethylation and concomitant silencing of MGMT also causes promoter silenc‐ing of other genes as well. In addition to silencing of genes, the CpG island methylatorphenotype can cause methylation of the promoter regions of long interspersed nuclear ele‐ment-1 (LINE-1) DNA sequences. Ohka et al. [139] point out that LINE-1 is a class of retro‐posons that are the most successful integrated mobile elements in the human genome, andaccount for about 18% of human DNA. Ohka et al. [139] found that LINE-1 methylation isdirectly proportional to MGMT promoter methylation in gliomas and suggested that LINE-1methylation could be used as a proxy to indicate the CpG island methylator phenotype sta‐tus in glioblastomas. This phenotype, likely associated with methylation of the MGMT pro‐moter, in turn, indicates whether treatment with the DNA alkylating agent temozolomidewill be beneficial in treatment of a patient with a glioblastoma, since MGMT removes thealkyl groups added to guanine by temozolomide.

DNA Damage, DNA Repair and Cancerhttp://dx.doi.org/10.5772/53919

435

Page 24: DNA Damage, DNA Repair and Cancer - InTech - Opencdn.intechopen.com/pdfs/43929/InTech-Dna_damage... · Chapter 16 DNA Damage, DNA Repair and Cancer Carol Bernstein, Anil R. Prasad,

14. Field defects

Field defects have been described in many types of gastrointestinal cancers [140]. A field de‐fect arises when an epimutation or mutation occurs in a stem cell that causes that stem cellto give rise to a number of daughter stem cells that can out-compete neighboring stem cells.These initial mutated cells form a patch of somewhat more rapidly growing cells (an initialfield defect). That patch then enlarges at the expense of neighboring cells, followed by, atsome point, an additional mutation or epimutation arising in one of the field defect stemcells so that this new stem cell with two advantageous mutations can generate daughterstem cells that can out-compete the surrounding field defect of cells that have just one ad‐vantageous mutation. As illustrated in Figure 4, this process of expanding sub-patches with‐in earlier patches will occur multiple times until a particular constellation of mutationsresults in a cancer (represented by the small dark patch in Figure 4. It should also be notedthat a cancer, once formed, continues to evolve and continues to produce sub clones. A renalcancer, sampled in 9 areas, had 40 ubiquitous mutations, 59 mutations shared by some, butnot all regions, and 29 “private” mutations only present in one region [141].

.

Figure 4. Schematic of a field defect in progression to cancer

New Research Directions in DNA Repair436

Page 25: DNA Damage, DNA Repair and Cancer - InTech - Opencdn.intechopen.com/pdfs/43929/InTech-Dna_damage... · Chapter 16 DNA Damage, DNA Repair and Cancer Carol Bernstein, Anil R. Prasad,

.

Figure 5. Colon resection including a colon cancer. Dashed arrows indicate grossly unremarkable colonic mucosa. Ul‐cerated hemorrhagic mass represents a moderately differentiated invasive adenocarcinoma. Solid arrow indicates theheaped up edge of the malignant ulcer

Figure 5 shows an opened resected segment of a human colon that has a colon cancer. Asillustrated by Bernstein et al. [142], there are about 100 colonic microscopic epithelial cryptsper sq mm in the colonic epithelium. The resection shown in Figure 5 has an area of about6.5 cm by 23 cm, or 150 sq cm, or 15, 000 sq mm. Thus this area has about 1.5 million crypts.There are 10-20 stem cells at the base of each colonic crypt [143, 144]. Therefore there arelikely about 15 million stem cells in the grossly unremarkable colonic mucosal epitheliumshown in Figure 5. Evidence reported by Facista et al. [112], and listed in Table 7, indicatesthat in many such resections, most of the stem cells in such an area up to 10 cm distant (ineach direction) from a colon cancer (such as in the grossly unremarkable area shown in Fig‐ure 5), and the majority of their differentiated daughter cells, are epigenetically deficient forprotein expression of the DNA repair genes ERCC1, PMS2 and/or XPF, although the epithe‐lium is histologically normal.

The stem cells most distant from the cancer, deficient for ERCC1, PMS2 and/or XPF, can beconsidered to constitute an outer ring, and be deficient as well in the inner rings, of a fielddefect schematically illustrated in Figure 4. The outer ring in Figure 4 includes, within itscircumscribed area, on the order of 15 million stem cells, presumably arising from an initialprogenitor stem cell deficient in DNA repair (due to epigenetic silencing). As a result of thisrepair deficit, the initial stem cell was genetically unstable, giving rise to an increased fre‐quency of mutations in its decendents. One daughter stem cell among its decendents had amutation that, by chance, provided a replicative advantage. This descendent then under‐went clonal expansion because of its replicative advantage. Among the further decendentsof the clone, new mutations arose frequently, since these descendents had a mutator pheno‐type [145], due to the repair deficiency passed down epigenetically from the original repair-defective stem cell. Among these new mutations, some would provide further replicativeadvantages, giving rise to a succession of more aggressively growing sub clones (innerrings), and eventually a cancer.

DNA Damage, DNA Repair and Cancerhttp://dx.doi.org/10.5772/53919

437

Page 26: DNA Damage, DNA Repair and Cancer - InTech - Opencdn.intechopen.com/pdfs/43929/InTech-Dna_damage... · Chapter 16 DNA Damage, DNA Repair and Cancer Carol Bernstein, Anil R. Prasad,

15. Exogenous carcinogenic agents cause reduced expression of DNArepair genes

Many known carcinogenic agents cause reduced expression of DNA repair genes or directlyinhibit the actions of DNA repair proteins. Table 8 lists examples of carcinogens that havesuch effects. Due to space limitations, many other such carcinogens are not listed. Thesefindings further link DNA damage to cancer.

Carcinogens Inhibit DNA Repair Gene Mechanism Shown Ref.

Arsenic compounds

PARP [146,147]

XRCC1

[146,148]Ligase 3

Ligase 4

DNA POLB, XRCC4[146]

DNA PKCS, TOPO2B

OGG1, ERCC1, XPF [149]

XPB, XPC, XPE [150]

P53Inhibition of P53 serine 15

phosphorylation[151]

Cadmium compounds

MSH2, ERCC1, XRCC1Promoter methylation [152]

OGG1

MSH2, MSH6 proteins Cd2+ binds to proteins [153]

OGG1 protein Oxidation of Ogg1 [154]

DNA-PK, XPD [155]

XPC [156]

Bile acids

deoxycholateMUTYH, OGG1 mRNA reduced [157]

BRCA1 [158]

lithocholate DNA POLB [159]

Lipid peroxidation

compounds

4-hydroxy-2-nonenal (4-HNE ) Nuc. Excision Repair NER protein adducts [160]

Malondialdehyde Nuc. Excision Repair NER protein adducts [161]

New Research Directions in DNA Repair438

Page 27: DNA Damage, DNA Repair and Cancer - InTech - Opencdn.intechopen.com/pdfs/43929/InTech-Dna_damage... · Chapter 16 DNA Damage, DNA Repair and Cancer Carol Bernstein, Anil R. Prasad,

Carcinogens Inhibit DNA Repair Gene Mechanism Shown Ref.

Oxidative stress

MisMatch RepairOxidative damage to MMR

proteins[162]

ERCC1 protein Oxidative attack [163]

OGG1 protein Degraded by calpain [164]

Gamma irrad. OGG1, XRCC1 mRNA reduced [165]

Benzo(a)pyrene BRCA1 miR-638 increased [166]

Methylcholanthrene/

diethylnitrosamineBRCA1, ERCC1, XRCC1, MLH1 [167]

Styrene XRCC1, OGG1, XPC mRNA reduced [168]

Aristolochic acid P53, PARP1, OGG1, ERCC1, MGMT mRNA reduced [169]

Antimony XPE mRNA reduced [170]

Nickel MGMT Promoter methylation [171]

Table 8. Examples of carcinogenic agents that cause reduced expression of DNA repair genes

16. Polyphenols can epigenetically increase expression of DNA repairgenes

Some polyphenols affect expression of many genes, including DNA repair genes, throughepigenetic alterations, as reviewed by Link et al. [172]. Examples of DNA repair genes ex‐pression increased by epigenetic alteration are listed in Table 9.

Phytochemical Plant source MechanismTargeted DNA

Repair GenesRef.

Epigalocatechin-3-gallate Green teaReversal of CpG island

methylationMGMT, MLH1 [173]

Dihydrocoumarin Yellow sweet clover p53 acetylation P53 [174]

Genistein SoyReversal of CpG island

methylationMGMT [173]

Genistein Soy Histone acetylation P53 [175]

Table 9. Examples of phytochemicals that increase expression of DNA repair genes by an epigenetic mechanism

DNA Damage, DNA Repair and Cancerhttp://dx.doi.org/10.5772/53919

439

Page 28: DNA Damage, DNA Repair and Cancer - InTech - Opencdn.intechopen.com/pdfs/43929/InTech-Dna_damage... · Chapter 16 DNA Damage, DNA Repair and Cancer Carol Bernstein, Anil R. Prasad,

17. Possible protection against cancer by phytochemicals that increaseDNA repair by unknown mechanisms

A recent review article by Collins et al. [176] summarizes some examples of micronutrients thataffect DNA repair gene expression, though by unknown mechanisms. Table 10 lists such phy‐tochemicals, without defined mechanisms, that increase DNA repair gene expression, alongwith commonly known foods that are high in those phytochemicals [177, 178, 179].

Phytochemical (test system) Examples of foods high in nutrient

Increased DNA

Repair Gene

Expression

Ref.

Ellagic acid (mice) Raspberries, pomeganateXPA, ERCC5, DNA

Ligase 3[180]

Silymarin (cells in vitro) Artichoke, milk thistle MGMT[181]

Curcumin (cells in vitro) Turmeric MGMT

Chlorogenic acid (cells in vitro)Blueberries, coffee, sunflower seeds,

artichokePARP

[182]

Caffeic acid (cells in vitro) coffee, cranberry, carrot PMS2

m-coumaric acid (cells in vitro) olives (and metabolite of caffeic acid) PARP, PMS2

3-(m-hydroxyphenyl) propionic acid (cells

in vitro)

(major metabolite of caffeic acid and

degradation product of proanthocyanidins

in chocholate)

PARP, PMS2

Table 10. Examples of phytochemicals that increase expression of DNA repair genes by unknown mechanisms

Bernstein et al. [182] evaluated antioxidants based on their ability to increase DNA repairproteins PARP-1 and Pms2 in vitro. They tested 19 anti-oxidant compounds and of these 19compounds only chlorogenic acid and its metabolic products: chlorogenic acid, caffeic acid,m-coumaric acid and 3-(m-hydroxyphenyl) propionic acid, increased expression of the twotested DNA repair genes in HCT-116 cells (Table 10).

Chlorogenic acid (CGA) (high in blueberries, coffee, sunflower seeds, artichoke) [177, 183,184] was then tested as a preventive agent in the recently devised diet-related mouse modelof colon cancer [60]. As described above in the section Exogenous DNA damaging agents incolorectal cancer, deoxycholic acid (DCA), a DNA damaging agent, at levels present after ahigh fat diet, can cause colorectal cancer. When DCA is added to the diet of wild-type miceto raise the level of DCA in the mouse feces to the level in feces of humans on a high fat diet,by 10 months of feeding 94% of the mice develop tumors in their colons with 56% develop‐ing colonic adenocarcinomas [60]. This mouse model develops tumors solely in the colon,phenotypically similar to development of colon cancer in humans. When CGA, equivalentto 3 cups of coffee a day for humans, was added to the DCA supplemented diet it was dra‐

New Research Directions in DNA Repair440

Page 29: DNA Damage, DNA Repair and Cancer - InTech - Opencdn.intechopen.com/pdfs/43929/InTech-Dna_damage... · Chapter 16 DNA Damage, DNA Repair and Cancer Carol Bernstein, Anil R. Prasad,

matically protective against development of colon cancer, reducing incidence of colon can‐cer significantly from 56% to 18% [60].

18. Targeting of chemotherapeutic agents to cancers deficient in DNArepair

As discussed above, DNA repair deficiency often arises early in progression to cancer andcan give rise to genomic instability, a general feature of cancers. If cancer cells are deficientin DNA repair they are likely to be more vulnerable than normal cells to inactivation byDNA damaging agents. This vulnerability of cancer cells can be exploited to the benefit ofthe patient. Some of the most clinically effective chemotherapeutic agents currently used incancer treatment are DNA damaging agents, and their therapeutic effectiveness appears tooften depend on deficient DNA repair in cancer cells.

In the next four sections we discuss repair deficiencies in cancer cells that can be effectivelytargeted by DNA damaging chemotherapeutic agents. In addition, deficiency in a DNA re‐pair pathway that arises during tumor development may make cancer cells more reliant ona remaining reduced set of DNA repair pathways for survival. Recent studies indicate thatdrugs that inhibit one of these alternative pathways in such cancers cells can be useful incancer therapy. Targeting cancer cells having a repair deficiency with specific DNA damag‐ing agents, or with agents that inhibit alternative repair pathways, offers a new promisingapproach for treating a variety of cancers.

19. Targeting cancers deficient in BRCA1

The BRCA1 (breast cancer 1 early onset) protein is employed in an important DNA repairpathway, homologous recombinational repair (HRR). This pathway removes a variety oftypes of DNA damages, and is the only pathway that can accurately remove double-stranddamages such as double-strand breaks and inter-strand cross-links. BRCA1 also has otherfunctions related to preservation of genome integrity (reviewed by Yun and Hiom [185]). In‐dividuals with a germ-line inherited defect in the BRCA1 gene are at increased risk of breast,ovarian and other cancers. In addition to inherited germ-line defects in BRCA1, deficienciesin expression of this gene may arise in somatic cells either by mutation or by epimutationduring progression to sporadic (non-germline) cancer.

Patients with a variety of types of cancer are treated effectively with chemotherapeuticagents that cause double-strand breaks (e.g. the topoisomerase inhibitor etoposide), or causeinter-strand cross-links (e.g. the platinum compound cisplatin). These damages can causecancer cells to undergo apoptosis (a form of cell death). However, patients treated withthese agents often prove to be intrinsically resistant, or develop resistance during treatment.Quinn et al. [186] demonstrated that BRCA1 expression is necessary for such resistance. Thisfinding suggests that BRCA1-mediated DNA repair can protect cancer cells from therapeutic

DNA Damage, DNA Repair and Cancerhttp://dx.doi.org/10.5772/53919

441

Page 30: DNA Damage, DNA Repair and Cancer - InTech - Opencdn.intechopen.com/pdfs/43929/InTech-Dna_damage... · Chapter 16 DNA Damage, DNA Repair and Cancer Carol Bernstein, Anil R. Prasad,

DNA damaging drugs. Thus, although high expression of BRCA1 may be initially beneficialto the individual by reducing the risk of developing cancer, it also may be detrimental oncecancer has developed by counteracting the therapeutic effect of DNA-damaging agents tar‐geted to the cancer cells.

Patients with non-small cell lung cancer (NSLC) are often treated with DNA cross-link‐ing platinum therapeutic compounds such as cisplatin, carboplatin or oxaliplatin. NSCLCis the leading cause of cancer deaths worldwide, and almost 70% of patients withNSCLC have locally advanced or metastatic disease at diagnosis. Improved survival afterplatinum-containing chemotherapy in metastatic NSCLC correlates with low BRCA1 ex‐pression in the primary tumor [187, 188]. This finding indicates that low BRCA1-mediat‐ed DNA repair is detrimental to the cancer upon treatment, and thus beneficial to thepatient. BRCA1 likely protects cancer cells by participating in a pathway that removesthe potentially lethal DNA cross-links introduced by the platinum drugs. Since lowBRCA1 expression in the tumor appears to be beneficial to the patient, Taron et al. [187]and Papadek et al. [188] concluded that BRCA1 expression is potentially an importanttool for use in cancer management and should be assessed for predicting chemosensitivi‐ty and tailoring chemotherapy in lung cancer.

Over 90% of ovarian cancers appear to arise sporadically in somatic cells and are associatedwith BRCA1 dysfunction. Weberpals et al. [189] showed for patients having sporadic ovari‐an cancer treated with platinum drugs, the median survival was longer for patients withlower expression of BRCA1 vs. higher BRCA1 expression (46 vs. 33 months).

20. Targeting cancers deficient in ERCC1

ERCC1 (Excision Repair Cross-Complementaion group 1) is a key protein needed to re‐move platinum adducts and repair inter- and intra-strand cross-links [190]. ERCC1 di‐merizes with XPF (xeroderma pigmentosum complementation group F) protein to form acomplex that can excise damaged DNA. Over-expression of ERCC1 is associated withcellular resistance to platinum compounds, whereas ERCC1 down-regulation sensitizescells to cisplatin [191, 192].

Cisplatin has made a major impact in the chemotherapeutic treatment of testicular cancer.Over 90% of patients with newly diagnosed testicular germ cell cancer, and 70 to 80% of pa‐tients with metastatic testicular cancer, can be cured using cisplatin based combination che‐motherapy [193]. Hypersensitivity of testicular cancer to cisplatin appears to be due to lowlevels of the three NER proteins ERCC1, XPF and XPA [194].

Simon et al. [195] evaluated ERCC1 mRNA expression in lung tumors as a predictor of sur‐vival of NSCLC patients. They found that patients with relatively low ERCC1 mRNA ex‐pression had poor overall survival. This finding suggests that low ERCC1-mediated DNArepair allows DNA damages to persist and give rise to carcinogenic mutations. However,they also noted that those NSCLC tumors with relatively low ERCC1 expression responded

New Research Directions in DNA Repair442

Page 31: DNA Damage, DNA Repair and Cancer - InTech - Opencdn.intechopen.com/pdfs/43929/InTech-Dna_damage... · Chapter 16 DNA Damage, DNA Repair and Cancer Carol Bernstein, Anil R. Prasad,

better to platinum based therapy. Lord et al. [196] found that low ERCC1 mRNA expressionin the primary tumor correlates with prolonged survival after cisplatin plus gemitabine che‐motherapy in NSCLC. Median overall survival with low ERCC1 expression tumors was 61.6weeks compared to 20.4 weeks for patients with high expression tumors.

Zhou et al. [197] reported that a particular genetic polymorphism that alters ERCC1 mRNAlevel predicts overall survival in advanced NSCLC patients treated with platinum basedchemotherapy. Olaussen et al. [198] found that patients with completely resected NSCLC tu‐mors that were ERCC1-negative benefited from adjuvant cisplatin-based chemotherapy,whereas patients with ERCC1-positive tumors did not benefit. They suggested that determi‐nation of ERCC1 expression in NSCLC cells before chemotherapy can make a contributionas an independent predictor of the effect of adjuvant chemotherapy. Papadaki et al. [188]found that ERCC1 mRNA level in the primary tumor of patients with metastatic NSCLCcould predict the effectiveness of cisplatin based chemotherapy. Low ERCC1 mRNA levelwas significantly associated with higher response rate, longer median progression-free sur‐vival and median overall survival. Leng et al. [199] found that patients with ERCC1 negativeexpression had a longer progression free survival and overall survival than ERCC1 positivepatients after receiving platinum based adjuvant therapy. Thus ERCC1 mRNA level, likeBRCA1 mRNA level (discussed above), in the primary tumor at the time of diagnosis couldbe used to predict platinum sensitivity of NSCLC.

ERCC1 expression also appears to have predictive significance for ovarian cancer. Dab‐holkar et al. [200] found in ovarian tumor tissues that ERCC1 mRNA expression levelswere higher in patients who were resistant to platinum based therapy than in those pa‐tients who responded to such therapy. Kang et al. [201] observed that a particular poly‐morphism of the ERCC1 gene sequence was associated with clinical outcome of platinumbased chemotherapy in patients with ovarian cancer. Weberpals et al. [189] also showedfor ovarian cancer patients that higher ERCC1 mRNA level, alone, or especially in com‐bination with higher BRCA1 mRNA level in the tumor, predicted shorter overall patientsurvival after platinum therapy.

ERCC1 protein expression is often reduced within colon cancers and in a field defect sur‐rounding these cancers [112]. For metastatic colorectal cancer patients receiving combinationoxaliplatin and fluorouracil chemotherapy, lower ERCC1 mRNA expression in the tumorpredicts longer survival [202]. Viguier et al. [203] found that a particular ERCC1 geneticpolymorphism predicts a better tumor response to oxaliplatin/5-fluorouracil combinationchemotherapy in patients with metastatic colorectal cancer.

Low ERCC1 mRNA levels also predict better response and survival for gastric cancer pa‐tients [204] and bladder cancer patients [205] receiving cisplatin-based chemotherapy.

Thus numerous studies involving cancer of the testis, lung, ovary, colon, stomach and blad‐der indicated that platinum based chemotherapy can enhance patient outcome when target‐ed specifically to tumors with low ERCC1 expression. Such tumors have diminished abilityto repair the DNA damages, particularly the cross-links, induced in the tumors by the plati‐num compound.

DNA Damage, DNA Repair and Cancerhttp://dx.doi.org/10.5772/53919

443

Page 32: DNA Damage, DNA Repair and Cancer - InTech - Opencdn.intechopen.com/pdfs/43929/InTech-Dna_damage... · Chapter 16 DNA Damage, DNA Repair and Cancer Carol Bernstein, Anil R. Prasad,

21. Targeting cancers deficient in MGMT

Alkylating agents, including chloroethylnitrosoureas, procarbazine and temozolomide, arecommonly used to treat malignant brain tumors. These agents cause DNA damage by add‐ing alkyl groups to DNA. Such damages may then be repaired or, if unrepaired, trigger celldeath. As an example, temozolomide methylates DNA at several sites generating mainly N7-methylguanine and N3-methyladenine adducts, which constitute nearly 90% of the totalmethylation events. However these adducts are efficiently removed and accurately replacedby the base excision repair pathway, and thus have low cytotoxic potential. About 5 to 10%of the methylation events caused by temozolomide produce O6-methylguanine which is cy‐totoxic, and this adduct accounts for the beneficial therapeutic effect of temozolomide andother alkylating agents on malignant brain tumors.

O6-methylguanine methyltransferase (MGMT) is a DNA repair enzyme that rapidly re‐verses alkylation (including methylation) at the O6 position of guanine, thus neutraliz‐ing the cytotoxic effects of chemotherapeutic alkylating agents such as temozolomide.High MGMT activity in tumor tissue is associated with resistance to alkylating agents.MGMT activity is controlled by a promoter sequence, and methylation of the CpG is‐land in the promoter silences the gene in cancer cells, so that these cells no longer pro‐duce MGMT. In addition, as described above, an increased level of miR-181d can alsodecrease MGMT expression and help the ability of temozolomide to give a beneficialtherapeutic effect [78].

Esteller et al. [206] showed that methylation of the MGMT promoter increases the re‐sponsiveness of the gliomas (brain tumors) to chemotherapeutic alkylating agents, lead‐ing to regression of the tumors and prolonged overall and disease free survival. Paz etal. [207] showed that hypermethylation of CpG islands within the promoter sequence ofthe MGMT gene predicts a better clinical response to temozolomide in primary gliomas.They considered that their results might open up possibilities for more customizedtreatments of human brain tumors. Hegi et al. [208] demonstrated a significantly im‐proved clinical outcome in patients with malignant glioma who had a methylatedMGMT promoter and were treated with temozolomide. The 18-month survival rate was62% among patients with a methylated MGMT promoter compared with only 8% in theabsence of promoter methylation. Hegi et al. [209] reviewed further evidence thatMGMT promoter methylation is associated with improved progression-free and overallsurvival in malignant glioma patients treated with alkylating agents. They also dis‐cussed strategies to overcome MGMT-mediated chemoresistance that are currently un‐der investigation. Upon reviewing the relevant evidence, Weller et al. [210] concludedthat MGMT promoter methylation is the key mechanism of MGMT gene silencing, andcould be used as a biomarker for predicting a favorable outcome in patients with ma‐lignant glioma who are exposed to alkylating chemotherapy. They considered that thisbiomarker is on the verge of entering clinical decision-making.

New Research Directions in DNA Repair444

Page 33: DNA Damage, DNA Repair and Cancer - InTech - Opencdn.intechopen.com/pdfs/43929/InTech-Dna_damage... · Chapter 16 DNA Damage, DNA Repair and Cancer Carol Bernstein, Anil R. Prasad,

22. Targeting cancers with a repair deficiency using a PARP inhibitor;synthetic lethality

If a tumor is deficient in an essential protein component of a DNA repair pathway, the can‐cer cells would likely be more reliant on remaining DNA repair pathways for survival.Drugs that inhibit one of these alternative pathways, in principle, might prove to be usefulin cancer therapy by selectively killing the cancer cells. An example of such an approach isthe use of poly(ADP-ribose) polymerase [PARP] inhibitors against tumors that are deficientin BRCA1 or BRCA2 [211]. This approach has provided proof-of-concept for an anticancerstrategy termed “synthetic lethality.” By this strategy the inhibition of a particular repairpathway in cancer cells that are already deficient in another repair pathway preferentiallyinduces greater toxicity in repair deficient cancer cells than in normal non-cancer cells. Cur‐rent research guided by this strategy is directed at finding new agents that inactivate proteincomponents of major repair pathways, and thus could be targeted against cancers that arealready deficient in another repair pathway [212].

A germ-line mutation in one BRCA1 or BRCA2 allele substantially increases the risk ofdeveloping several cancers, including breast, ovarian, and prostate cancer. Diploid cellsheterozygous for either a BRCA1 or a BRCA2 mutant allele may lose expression of theremaining wild-type allele, resulting in deficient homologous recombinational repair.This loss causes an increase in unrepaired DSBs that can lead to mutations (throughcompensatory inaccurate repair) and chromosomal aberrations that drive carcinogenesis.Inactivation of the wild-type allele in the cell lineage leading to the tumor is thought tobe an obligate step in this carcinogenesis pathway, a step that does not occur in thenormal non-cancer tissues of the patient.

The deficiency in homologous recombinational repair is thus specific to the tumor, and canbe exploited by employing PARP inhibitors. Ordinarily, single-strand breaks (SSBs), as dis‐tinct from DSBs, are repaired by the base excision repair pathway, in which the enzymePARP1 plays a key role. The inhibition of PARP1 leads to the accumulation of DNA SSBs.Unrepaired SSBs can give rise to DSBs at replication forks during DNA replication. ThusPARP inhibition in tumor cells with deficient homologous recombinational repair (becauseof the absence of BRCA1 or BRCA2) generates unrepaired SSBs that are likely to cause anoverwhelming accumulation of DSBs leading to tumor cell death. In contrast, the normal tis‐sues of a patient consists of cells that are heterozygous for a BRCA1 or BRCA2 mutant alleleand therefore retain homologous recombinational repair function, and have a sensitivity toPARP inhibitors similar to that of wild-type cells. Thus PARP inhibition induces selective tu‐mor cell killing while sparing normal cells.

Fong et al. [213] conducted a preliminary clinical evaluation of the oral PARP inhibitorolaparib. They observed that 63% of patients carrying BRCA1 or BRCA2 mutations whohad ovarian, breast or prostate cancer had a clinical benefit from treatment with olapar‐ib with few adverse side effects. This is an example of the concept of “synthetic lethali‐ty” which occurs when there is a potent lethal synergy between two otherwise non-

DNA Damage, DNA Repair and Cancerhttp://dx.doi.org/10.5772/53919

445

Page 34: DNA Damage, DNA Repair and Cancer - InTech - Opencdn.intechopen.com/pdfs/43929/InTech-Dna_damage... · Chapter 16 DNA Damage, DNA Repair and Cancer Carol Bernstein, Anil R. Prasad,

lethal events. The two events in this case are (1) a specific PARP inhibitor blocks repairof SSBs causing an increase in SSBs leading to an increase in DSBs; and (2) a tumor re‐stricted genetic loss of function or homologous recombinational repair that is ordinarilyneeded to accurately repair these DSBs.

A subsequent trial of olaparib in BRCA mutation-associated breast cancer demonstrated ob‐jective positive response rates of 41%, again with limited toxicity [214]. About 10% of wom‐en with ovarian cancer carry a BRCA1 or BRCA2 mutant allele. Audeh et al. [215] showedthat the oral PARP inhibitor olaparib has antitumor activity in women carriers of BRCA1 orBRCA2 alleles who have ovarian cancer. The objective positive response rate was 33%.

23. Overview of the role of DNA damage and repair in carcinogenesis

In this section we present a brief overview of the relationship of DNA damage and re‐pair to carcinogenesis, and the implications of this relationship for strategies of preven‐tion and therapy, emphasizing the evidence reviewed above. Carcinogenesis is generallyviewed as a Darwinian process that occurs in a somatic cell lineage by mutation or epi‐mutation and natural selection. Natural selection operates on the basis of the adaptivebenefit to individual cells in the lineage of more rapid cell division or higher resistanceto cell death (apoptosis) than occurs in neighboring cells. Most of the random mutationsand epimutations that arise during progression to cancer are likely to be disadvanta‐geous or neutral from the prospective of the emerging cancerous cells, and only thosethat promote more rapid overall growth are advantageous. The cell lineage that ultimate‐ly becomes a cancer probably passes through a series of evolutionary pre-cancerousstages involving sequential rounds of mutation/epimutation and selection [216]. The ini‐tial stage is probably a lineage of cells with a small selective advantage that forms anearly field within a tissue. Within this defective field successive mutation and selectionevents occur which finally give rise to an invasive and then metastatic cell lineage. Dur‐ing this process the cell lineage acquires the hallmarks of cancer (summarized by Hana‐han and Weinberg [217]). These include: sustaining proliferative signaling, evadinggrowth suppressors, resisting cell death, enabling replicative immortality, inducing angio‐genesis, reprogramming energy metabolism, and evading immune destruction.

Mutations arise from unrepaired DNA damages, either by translesion synthesis duringDNA replication or by inaccurate repair of DNA damages, as in the inaccurate process ofnon-homologous end joining of double-strand breaks. Mutations may also arise by sponta‐neous replication errors without the intervention of DNA damage, but this source of muta‐tion is likely less frequent than mutations caused by DNA damage. The primary cause(s) ofepimutations (such as CpG island methylations) are not well understood, but evidence sug‐gests that epimutations arise during the repair processes that remove DNA damages. Thesources of DNA damage underlying carcinogenesis can be extrinsic or intrinsic. Epidemio‐logic evidence suggests that a large proportion of the DNA damages contributing to cancerarise from extrinsic stressful conditions, including such factors as smoking, high fat diet, cer‐

New Research Directions in DNA Repair446

Page 35: DNA Damage, DNA Repair and Cancer - InTech - Opencdn.intechopen.com/pdfs/43929/InTech-Dna_damage... · Chapter 16 DNA Damage, DNA Repair and Cancer Carol Bernstein, Anil R. Prasad,

tain infections and UV light exposure. The possible contribution from intrinsic causes, suchas free radical production during normal metabolism, have not been assessed. A pervasivecharacteristic of human tumors is genomic instability [217]. A likely major source of this in‐stability is loss of DNA repair capability. Germ line mutations in DNA repair genes general‐ly lead to syndromes characterized by a greatly increased risk of cancer. The majority ofcancers arise sporadically, i.e. are not primarily due to germ line mutations. A frequent char‐acteristic of sporadic cancers is loss of expression of one or more DNA repair proteinsthrough epigenetic silencing. The several different DNA repair pathways that occur in mam‐malian cells each specialize in removing different types of damage, but they are also partial‐ly overlapping. Thus reduction of a particular repair pathway may have differentcarcinogenic consequences from loss of another repair pathway [218]. However, the deleteri‐ous effect of loss of one pathway may be partially ameliorated by another functioning path‐way.

This general view of the role of DNA damage and repair in carcinogenesis has implicationsfor the prevention and treatment of cancer. Cancer incidence could be substantially reducedby a general avoidance of the known sources of DNA damage such as smoking. In additionto avoiding DNA damage, it should also be beneficial to increase DNA repair, or at least toavoid extrinsic factors that decrease repair. The factors affecting repair capability are lesswell studied than those causing DNA damage, but several are known, and a significant ben‐efit may be derived from considering such factors as well.

The finding that DNA repair deficiency is a common feature of cancers, and is perhaps theunderlying cause of the genetic instability of cancers, has implications for therapy. If a can‐cer is composed of cells deficient in DNA repair, it is, in principle, vulnerable to agents thatcause DNA damage. Thus a chemotherapeutic DNA damaging agent can be targeted to can‐cers that lack the capability to repair the particular type of DNA damage caused by theagent. This can lead to a level of DNA damages that overwhelms the defenses of the cancercells and causes their death. Non-cancerous cells with normal repair would not be targeted.Thus the toxicity of such DNA damaging agents to the treated patient would be limited. Adramatic example of such targeted therapy is the high cure rate of testicular cancer due to adefect in the ability of the cancer cells to repair DNA inter-strand cross-links, and the use ofcross-linking platinum compounds to kill such cells.

Another strategy, which is currently the basis for numerous ongoing clinical trials, involvessynthetic lethality. By this strategy cancers that are deficient in one DNA repair pathway canbe made more vulnerable to DNA damage by treatment with agents that inhibit an addi‐tional repair pathway. Promising clinical results, so far, have been obtained in the treatmentof patients with breast and ovarian cancer due to an inherited genetic defect in the homolo‐gous recombinational repair pathway. Such cancers are deficient in the ability to repair dou‐ble-strand breaks. Treatment of these cancers with an agent that interferes with anotherpathway that ordinarily repairs single-strand breaks allows such breaks to accumulate andto be converted to double-strand breaks during DNA replication. The increase in double-strand breaks appears to overwhelm the cancer cells, while sparing normal cells, thus pro‐viding positive clinical benefit to the patient without much toxicity.

DNA Damage, DNA Repair and Cancerhttp://dx.doi.org/10.5772/53919

447

Page 36: DNA Damage, DNA Repair and Cancer - InTech - Opencdn.intechopen.com/pdfs/43929/InTech-Dna_damage... · Chapter 16 DNA Damage, DNA Repair and Cancer Carol Bernstein, Anil R. Prasad,

Author details

Carol Bernstein1*, Anil R. Prasad2, Valentine Nfonsam3 and Harris Bernstein4

*Address all correspondence to: [email protected]

1 Research Service Line, Southern Arizona Veterans Affairs Health Care System, Tucson,AZ, USA

2 Department of Pathology, University of Arizona, Tucson, AZ, USA

3 Department of Surgery, University of Arizona, Tucson, AZ, USA

4 Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, USA

References

[1] Gregg SQ, Robinson AR, Niedernhofer LJ. Physiological consequences of defects inERCC1-XPF DNA repair endonuclease. DNA Repair (Amst) 2011;10(7) 781-791.

[2] Agrelo R, Cheng WH, Setien F, Ropero S, Espada J, Fraga MF, Herranz M, Paz MF,Sanchez-Cespedes M, Artiga J, Guerrero D, Castells A, von Kobbe C, Bohr VA, Estel‐ler M. Epigenetic inactivation of the premature aging Werner syndrome gene in hu‐man cancer. Proc Natl Acad Sci U S A 2006;103(23) 8822-8827.

[3] Dreesen O, Stewart CL. Accelerated aging syndromes, are they relevant to normalhuman aging? Aging (Albany NY) 2011;3(9) 889-895.

[4] Wood LD, Parsons DW, Jones S, Lin J, Sjöblom T, Leary RJ, Shen D, Boca SM, BarberT, Ptak J, Silliman N, Szabo S, Dezso Z, Ustyanksky V, Nikolskaya T, Nikolsky Y,Karchin R, Wilson PA, Kaminker JS, Zhang Z, Croshaw R, Willis J, Dawson D, Shipit‐sin M, Willson JK, Sukumar S, Polyak K, Park BH, Pethiyagoda CL, Pant PV, Ballin‐ger DG, Sparks AB, Hartigan J, Smith DR, Suh E, Papadopoulos N, Buckhaults P,Markowitz SD, Parmigiani G, Kinzler KW, Velculescu VE, Vogelstein B. The genomiclandscapes of human breast and colorectal cancers. Science 2007;318(5853) 1108-1113.

[5] Cancer Genome Atlas Network. Comprehensive molecular characterization of hu‐man colon and rectal cancer. Nature 2012;487(7407) 330-337. doi: 10.1038/nature11252.

[6] Leary RJ, Lin JC, Cummins J, Boca S, Wood LD, Parsons DW, Jones S, Sjöblom T,Park BH, Parsons R, Willis J, Dawson D, Willson JK, Nikolskaya T, Nikolsky Y, Ko‐pelovich L, Papadopoulos N, Pennacchio LA, Wang TL, Markowitz SD, ParmigianiG, Kinzler KW, Vogelstein B, Velculescu VE. Integrated analysis of homozygous de‐letions, focal amplifications, and sequence alterations in breast and colorectal can‐cers. Proc Natl Acad Sci USA 2008;105(42) 16224-16229.

New Research Directions in DNA Repair448

Page 37: DNA Damage, DNA Repair and Cancer - InTech - Opencdn.intechopen.com/pdfs/43929/InTech-Dna_damage... · Chapter 16 DNA Damage, DNA Repair and Cancer Carol Bernstein, Anil R. Prasad,

[7] Schnekenburger M, Diederich M. Epigenetics Offer New Horizons for ColorectalCancer Prevention. Curr Colorectal Cancer Rep 2012;8(1) 66-81.

[8] O'Hagan HM, Mohammad HP, Baylin SB. Double strand breaks can initiate gene si‐lencing and SIRT1-dependent onset of DNA methylation in an exogenous promoterCpG island. PLoS Genet 2008;4(8) e1000155.

[9] Malanga M, Althaus FR. The role of poly(ADP-ribose) in the DNA damage signalingnetwork. Biochem Cell Biol 2005;83(3) 354-64.

[10] Gottschalk AJ, Timinszky G, Kong SE, Jin J, Cai Y, Swanson SK, Washburn MP, Flo‐rens L, Ladurner AG, Conaway JW, Conaway RC. Poly(ADP-ribosyl)ation directs re‐cruitment and activation of an ATP-dependent chromatin remodeler. Proc Natl AcadSci U S A 2009;106(33) 13770-13774.

[11] Lin JC, Jeong S, Liang G, Takai D, Fatemi M, Tsai YC, Egger G, Gal-Yam EN, JonesPA. Role of nucleosomal occupancy in the epigenetic silencing of the MLH1 CpG is‐land. Cancer Cell 2007;12(5) 432-444.

[12] Tabish AM, Poels K, Hoet P, Godderis L. Epigenetic Factors in Cancer Risk: Effect ofChemical Carcinogens on Global DNA Methylation Pattern in Human TK6 Cells.PLoS One 2012;7(4) e34674. Epub 2012 Apr 11.

[13] Stefanska B, Karlic H, Varga F, Fabianowska-Majewska K, Haslberger AG. Epigenet‐ic mechanisms in anti-cancer actions of bioactive food components-the implicationsin cancer prevention. Br J Pharmacol 2012 Apr 27. doi: 10.1111/j.1476-5381.2012.02002.x. [Epub ahead of print]

[14] Jacinto FV, Esteller M. Mutator pathways unleashed by epigenetic silencing in hu‐man cancer. Mutagenesis 2007;22(4) 247-253.

[15] De Bont R, van Larebeke N. Endogenous DNA damage in humans: a review of quan‐titative data. Mutagenesis 2004;19(3) 169-185. http://mutage.oxfordjournals.org/content/19/3/169.full.pdf+html

[16] Helbock HJ, Beckman KB, Shigenaga MK, Walter PB, Woodall AA, Yeo HC, AmesBN. DNA oxidation matters: the HPLC-electrochemical detection assay of 8-oxo-de‐oxyguanosine and 8-oxo-guanine. Proc Natl Acad Sci U S A. 1998;95(1) 288-293.

[17] Nakamura J, Swenberg JA. Endogenous apurinic/apyrimidinic sites in genomic DNAof mammalian tissues. Cancer Res 1999;59(11) 2522-2526.

[18] Fraga CG, Shigenaga MK, Park JW, Degan P, Ames BN. Oxidative damage to DNAduring aging: 8-hydroxy-2'-deoxyguanosine in rat organ DNA and urine. Proc NatlAcad Sci U S A 1990;87(12) 4533-4537.

[19] Ames BN, Shigenaga MK, Hagen TM. Oxidants, antioxidants, and the degenerativediseases of aging. Proc Natl Acad Sci U S A 1993;90(17) 7915-7922.

[20] Foksinski M, Rozalski R, Guz J, Ruszkowska B, Sztukowska P, Piwowarski M,Klungland A, Olinski R. Urinary excretion of DNA repair products correlates with

DNA Damage, DNA Repair and Cancerhttp://dx.doi.org/10.5772/53919

449

Page 38: DNA Damage, DNA Repair and Cancer - InTech - Opencdn.intechopen.com/pdfs/43929/InTech-Dna_damage... · Chapter 16 DNA Damage, DNA Repair and Cancer Carol Bernstein, Anil R. Prasad,

metabolic rates as well as with maximum life spans of different mammalian species.Free Radic Biol Med 2004;37(9) 1449-1454.

[21] Lindahl T. DNA repair enzymes acting on spontaneous lesions in DNA. In: NicholsWW and Murphy DG (eds.) DNA Repair Processes. Symposia Specialists, Miami,1977. p225-240.

[22] Tice, R.R., and Setlow, R.B. DNA repair and replication in aging organisms and cells.In: Finch EE and Schneider EL (eds.) Handbook of the Biology of Aging. Van Nos‐trand Reinhold, New York. 1985. p173-224.

[23] Lindahl T, Nyberg B. Rate of depurination of native deoxyribonucleic acid. Biochem‐istry 1972;11(19) 3610-3618.

[24] Lindahl T. Instability and decay of the primary structure of DNA. Nature1993;362(6422) 709-715.

[25] Nakamura J, Walker VE, Upton PB, Chiang SY, Kow YW, Swenberg JA. Highly sen‐sitive apurinic/apyrimidinic site assay can detect spontaneous and chemically in‐duced depurination under physiological conditions. Cancer Res 1998;58(2) 222-225.

[26] Haber JE. DNA recombination: the replication connection. Trends Biochem Sci1999;24(7) 271-275.

[27] Vilenchik MM, Knudson AG. Endogenous DNA double-strand breaks: production,fidelity of repair, and induction of cancer. Proc Natl Acad Sci U S A 2003;100(22)12871-12876. http://www.ncbi.nlm.nih.gov/pmc/articles/PMC240711/?tool=pubmed

[28] Nam EA, Cortez D. ATR signalling: more than meeting at the fork. Biochem J2011;436(3) 527-536.

[29] Maher RL, Branagan AM, Morrical SW. Coordination of DNA replication and recom‐bination activities in the maintenance of genome stability. J Cell Biochem2011;112(10) 2672-82. doi: 10.1002/jcb.23211.

[30] Holmes GE, Bernstein C, Bernstein H. Oxidative and other DNA damages as the ba‐sis of aging: a review. Mutat Res 1992;275(3-6) 305-315.

[31] Hoeijmakers JH. DNA damage, aging, and cancer. N Engl J Med 2009;361(15)1475-1485.

[32] Ou J, DeLany JP, Zhang M, Sharma S, O'Keefe SJ. Association between low colonicshort-chain fatty acids and high bile acids in high colon cancer risk populations. NutrCancer 2012;64(1) 34-40.

[33] O'Keefe SJ, Kidd M, Espitalier-Noel G, Owira P. Rarity of colon cancer in Africans isassociated with low animal product consumption, not fiber. Am J Gastroenterol1999;94(5) 1373-1380.

[34] Maskarinec G, Noh JJ. The effect of migration on cancer incidence among Japanese inHawaii. Ethn Dis 2004 Summer;14(3) 431-439.

New Research Directions in DNA Repair450

Page 39: DNA Damage, DNA Repair and Cancer - InTech - Opencdn.intechopen.com/pdfs/43929/InTech-Dna_damage... · Chapter 16 DNA Damage, DNA Repair and Cancer Carol Bernstein, Anil R. Prasad,

[35] Jemal A, Bray F, Center MM, Ferlay J, Ward E, Forman D. Global cancer statistics.CA Cancer J Clin 2011;61(2) 69-90.

[36] Alberg AJ, Ford JG, Samet JM; American College of Chest Physicians. Epidemiologyof lung cancer: ACCP evidence-based clinical practice guidelines (2nd edition). Chest2007;132(3 Suppl) 29S-55S.

[37] Cunningham FH, Fiebelkorn S, Johnson M, Meredith C. A novel application of theMargin of Exposure approach: segregation of tobacco smoke toxicants. Food ChemToxicol 2011;49(11) 2921-2933.

[38] Liu XY, Zhu MX, Xie JP. Mutagenicity of acrolein and acrolein-induced DNA ad‐ducts. Toxicol Mech Methods 2010;20(1) 36-44.

[39] Alexandrov K, Rojas M, Satarug S. The critical DNA damage by benzo(a)pyrene inlung tissues of smokers and approaches to preventing its formation. Toxicol Lett2010;198(1) 63-68.

[40] Speit G, Merk O. Evaluation of mutagenic effects of formaldehyde in vitro: detectionof crosslinks and mutations in mouse lymphoma cells. Mutagenesis 2002;17(3)183-187.

[41] Grogan D, Jinks-Robertson S. Formaldehyde-induced mutagenesis in Saccharomycescerevisiae: molecular properties and the roles of repair and bypass systems. MutatRes 2012;731(1-2) 92-98.

[42] Pu X, Kamendulis LM, Klaunig JE. Acrylonitrile-induced oxidative stress and oxida‐tive DNA damage in male Sprague-Dawley rats. Toxicol Sci 2009;111(1) 64-71.

[43] Marnett LJ. Oxy radicals, lipid peroxidation and DNA damage. Toxicology2002;181-182 219-222.

[44] Koturbash I, Scherhag A, Sorrentino J, Sexton K, Bodnar W, Swenberg JA, Beland FA,Pardo-Manuel Devillena F, Rusyn I, Pogribny IP. Epigenetic mechanisms of mouseinterstrain variability in genotoxicity of the environmental toxicant 1, 3-butadiene.Toxicol Sci. 2011;122(2) 448-456.

[45] Garcia CC, Angeli JP, Freitas FP, Gomes OF, de Oliveira TF, Loureiro AP, Di MascioP, Medeiros MH. (13C2)-Acetaldehyde promotes unequivocal formation of 1, N2-propano-2'-deoxyguanosine in human cells. J Am Chem Soc 2011;133(24) 9140-9143.

[46] Tompkins EM, McLuckie KI, Jones DJ, Farmer PB, Brown K. Mutagenicity of DNAadducts derived from ethylene oxide exposure in the pSP189 shuttle vector replicat‐ed in human Ad293 cells. Mutat Res 2009;678(2) 129-137.

[47] Gelmont D, Stein RA, Mead JF. Isoprene-the main hydrocarbon in human breath. Bi‐ochem Biophys Res Commun 1981;99(4) 1456-1460.

[48] Senthilmohan ST, McEwan MJ, Wilson PF, Milligan DB, Freeman CG. Real time anal‐ysis of breath volatiles using SIFT-MS in cigarette smoking. Redox Rep 2001;6(3)185-187.

DNA Damage, DNA Repair and Cancerhttp://dx.doi.org/10.5772/53919

451

Page 40: DNA Damage, DNA Repair and Cancer - InTech - Opencdn.intechopen.com/pdfs/43929/InTech-Dna_damage... · Chapter 16 DNA Damage, DNA Repair and Cancer Carol Bernstein, Anil R. Prasad,

[49] Fabiani R, Rosignoli P, De Bartolomeo A, Fuccelli R, Morozzi G. DNA-damagingability of isoprene and isoprene mono-epoxide (EPOX I) in human cells evaluatedwith the comet assay. Mutat Res 2007;629(1) 7-13.

[50] Phillips DH. Smoking-related DNA and protein adducts in human tissues. Carcino‐genesis 2002;23(12) 1979-2004.

[51] Giovannucci E, Martínez ME. Tobacco, colorectal cancer, and adenomas: a review ofthe evidence. J Natl Cancer Inst 1996;88(23) 1717-1730.

[52] de Kok TM, van Maanen JM. Evaluation of fecal mutagenicity and colorectal cancerrisk. Mutat Res 2000; 463(1) 53-101.

[53] Pearson JR, Gill CI, Rowland IR. Diet, fecal water, and colon cancer--development ofa biomarker. Nutr Rev 2009;67(9) 509-526.

[54] Bernstein H, Bernstein C, Payne CM, Dvorak K. Bile acids as endogenous etiologicagents in gastrointestinal cancer. World J Gastroenterol 2009;15(27) 3329-3340.

[55] Payne CM, Crowley-Skillicorn C, Bernstein C, Holubec H, Moyer MP, Bernstein H.Hydrophobic bile acid-induced micronuclei formation, mitotic perturbations, anddecreases in spindle checkpoint proteins: relevance to genomic instability in coloncarcinogenesis. Nutr Cancer 2010;62(6) 825-840.

[56] Bernstein H, Bernstein C, Payne CM, Dvorakova K, Garewal H. Bile acids as carcino‐gens in human gastrointestinal cancers. Mutat Res 2005;589(1) 47-65.

[57] Hursting SD, Thornquist M, Henderson MM. Types of dietary fat and the incidenceof cancer at five sites. Prev Med 1990;19(3) 242-253.

[58] Reddy BS, Hanson D, Mangat S, Mathews L, Sbaschnig M, Sharma C, Simi B. Effectof high-fat, high-beef diet and of mode of cooking of beef in the diet on fecal bacterialenzymes and fecal bile acids and neutral sterols. J Nutr 1980;110(9) 1880-1887.

[59] Stadler J, Stern HS, Yeung KS, McGuire V, Furrer R, Marcon N, Bruce WR. Effect ofhigh fat consumption on cell proliferation activity of colorectal mucosa and on solu‐ble faecal bile acids. Gut 1988;29(10) 1326-1331.

[60] Bernstein C, Holubec H, Bhattacharyya AK, Nguyen H, Payne CM, Zaitlin B, Bern‐stein H. Carcinogenicity of deoxycholate, a secondary bile acid. Arch Toxicol2011;85(8) 863-871

[61] Handa O, Naito Y, Yoshikawa T. Redox biology and gastric carcinogenesis: the roleof Helicobacter pylori. Redox Rep 2011;16(1) 1-7.

[62] Wei L, Gravitt PE, Song H, Maldonado AM, Ozbun MA. Nitric oxide induces earlyviral transcription coincident with increased DNA damage and mutation rates in hu‐man papillomavirus-infected cells. Cancer Res 2009;69(11) 4878-4884.

New Research Directions in DNA Repair452

Page 41: DNA Damage, DNA Repair and Cancer - InTech - Opencdn.intechopen.com/pdfs/43929/InTech-Dna_damage... · Chapter 16 DNA Damage, DNA Repair and Cancer Carol Bernstein, Anil R. Prasad,

[63] Goldman A, Shahidullah M, Goldman D, Khailova L, Watts G, Delamere N, DvorakK. A novel mechanism of acid and bile acid-induced DNA damage involving Na+/H+ exchanger: implication for Barrett's oesophagus. Gut 2010;59(12) 1606-1616.

[64] Smela ME, Hamm ML, Henderson PT, Harris CM, Harris TM, Essigmann JM. Theaflatoxin B(1) formamidopyrimidine adduct plays a major role in causing the typesof mutations observed in human hepatocellular carcinoma. Proc Natl Acad Sci U S A2002;99(10) 6655-6660.

[65] Kanavy HE, Gerstenblith MR. Ultraviolet radiation and melanoma. Semin CutanMed Surg 2011;30(4) 222-228

[66] Lichtenstein P, Holm NV, Verkasalo PK, Iliadou A, Kaprio J, Koskenvuo M, PukkalaE, Skytthe A, Hemminki K. Environmental and heritable factors in the causation ofcancer--analyses of cohorts of twins from Sweden, Denmark, and Finland. N Engl JMed 2000;343(2) 78-85.

[67] https://dnapittcrew.upmc.com/db/orthologs.php

[68] http://sciencepark.mdanderson.org/labs/wood/dna_repair_genes.html

[69] Wood RD, Mitchell M, Sgouros J, Lindahl T. Human DNA repair genes. Science2001;291(5507) 1284-1289.

[70] Wood RD, Mitchell M, Lindahl T. Human DNA repair genes, 2005. Mutat Res2005;577(1-2) 275-283.

[71] Bernstein C, Bernstein H, Payne CM, Garewal H. DNA repair/pro-apoptotic dual-role proteins in five major DNA repair pathways: fail-safe protection against carcino‐genesis. Mutat Res 2002;511(2) 145-178.

[72] Foulkes WD. Inherited susceptibility to common cancers. N Engl J Med 2008;359(20)2143-2153.

[73] O'Hagan HM, Wang W, Sen S, Destefano Shields C, Lee SS, Zhang YW, ClementsEG, Cai Y, Van Neste L, Easwaran H, Casero RA, Sears CL, Baylin SB. Oxidativedamage targets complexes containing DNA methyltransferases, SIRT1, and poly‐comb members to promoter CpG Islands. Cancer Cell. 2011;20(5) 606-619.

[74] Malkin D. Li-fraumeni syndrome. Genes Cancer 2011;2(4) 475-484.

[75] Roukos DH. Genome-wide association studies: how predictable is a person's cancerrisk? Expert Rev Anticancer Ther 2009;9(4) 389-392.

[76] Truninger K, Menigatti M, Luz J, Russell A, Haider R, Gebbers JO, Bannwart F,Yurtsever H, Neuweiler J, Riehle HM, Cattaruzza MS, Heinimann K, Schär P, JiricnyJ, Marra G. Immunohistochemical analysis reveals high frequency of PMS2 defects incolorectal cancer. Gastroenterology 2005;128(5) 1160-1171.

[77] Valeri N, Gasparini P, Fabbri M, Braconi C, Veronese A, Lovat F, Adair B, Vannini I,Fanini F, Bottoni A, Costinean S, Sandhu SK, Nuovo GJ, Alder H, Gafa R, Calore F,

DNA Damage, DNA Repair and Cancerhttp://dx.doi.org/10.5772/53919

453

Page 42: DNA Damage, DNA Repair and Cancer - InTech - Opencdn.intechopen.com/pdfs/43929/InTech-Dna_damage... · Chapter 16 DNA Damage, DNA Repair and Cancer Carol Bernstein, Anil R. Prasad,

Ferracin M, Lanza G, Volinia S, Negrini M, McIlhatton MA, Amadori D, Fishel R,Croce CM. Modulation of mismatch repair and genomic stability by miR-155. ProcNatl Acad Sci U S A 2010;107(15) 6982-6987.

[78] Zhang W, Zhang J, Hoadley K, Kushwaha D, Ramakrishnan V, Li S, Kang C, You Y,Jiang C, Song SW, Jiang T, Chen CC. miR-181d: a predictive glioblastoma biomarkerthat downregulates MGMT expression. Neuro Oncol 2012;14(6) 712-719.

[79] Spiegl-Kreinecker S, Pirker C, Filipits M, Lötsch D, Buchroithner J, Pichler J, Silye R,Weis S, Micksche M, Fischer J, Berger W.O6-Methylguanine DNA methyltransferaseprotein expression in tumor cells predicts outcome of temozolomide therapy in glio‐blastoma patients. Neuro Oncol 2010 Jan;12(1) 28-36.

[80] Lahtz C, Pfeifer GP. Epigenetic changes of DNA repair genes in cancer. J Mol CellBiol 2011;3(1) 51-58.

[81] Narayanan L, Fritzell JA, Baker SM, Liskay RM, Glazer PM. Elevated levels of muta‐tion in multiple tissues of mice deficient in the DNA mismatch repair gene Pms2.Proc Natl Acad Sci U S A 1997;94(7) 3122-3127.

[82] Hegan DC, Narayanan L, Jirik FR, Edelmann W, Liskay RM, Glazer PM. Differingpatterns of genetic instability in mice deficient in the mismatch repair genes Pms2,Mlh1, Msh2, Msh3 and Msh6. Carcinogenesis 2006;27(12) 2402-2408.

[83] Tutt AN, van Oostrom CT, Ross GM, van Steeg H, Ashworth A. Disruption of Brca2increases the spontaneous mutation rate in vivo: synergism with ionizing radiation.EMBO Rep 2002;3(3) 255-260.

[84] German J. Bloom's syndrome. I. Genetical and clinical observations in the first twen‐ty-seven patients. Am J Hum Genet 1969;21(2) 196-227.

[85] Nagaraju G, Scully R. Minding the gap: the underground functions of BRCA1 andBRCA2 at stalled replication forks. DNA Repair (Amst) 2007;6(7) 1018-1031.

[86] Lancaster JM, Powell CB, Kauff ND, Cass I, Chen LM, Lu KH, Mutch DG, BerchuckA, Karlan BY, Herzog TJ; Society of Gynecologic Oncologists Education Committee.Society of Gynecologic Oncologists Education Committee statement on risk assess‐ment for inherited gynecologic cancer predispositions. Gynecol Oncol 2007;107(2)159-162.

[87] Keimling M, Volcic M, Csernok A, Wieland B, Dörk T, Wiesmüller L. Functionalcharacterization connects individual patient mutations in ataxia telangiectasia mutat‐ed (ATM) with dysfunction of specific DNA double-strand break-repair signalingpathways. FASEB J 2011;25(11) 3849-3860.

[88] Thompson LH, Schild D. Recombinational DNA repair and human disease. MutatRes 2002;509(1-2) 49-78.

[89] Chrzanowska KH, Gregorek H, Dembowska-Bagińska B, Kalina MA, Digweed M.Nijmegen breakage syndrome (NBS). Orphanet J Rare Dis 2012;7 13.

New Research Directions in DNA Repair454

Page 43: DNA Damage, DNA Repair and Cancer - InTech - Opencdn.intechopen.com/pdfs/43929/InTech-Dna_damage... · Chapter 16 DNA Damage, DNA Repair and Cancer Carol Bernstein, Anil R. Prasad,

[90] Rapp A, Greulich KO. After double-strand break induction by UV-A, homologousrecombination and nonhomologous end joining cooperate at the same DSB if bothsystems are available. J Cell Sci 2004;117(Pt 21) 4935-4945.

[91] Bartkova J, Tommiska J, Oplustilova L, Aaltonen K, Tamminen A, Heikkinen T, Mis‐trik M, Aittomäki K, Blomqvist C, Heikkilä P, Lukas J, Nevanlinna H, Bartek J. Aber‐rations of the MRE11-RAD50-NBS1 DNA damage sensor complex in human breastcancer: MRE11 as a candidate familial cancer-predisposing gene. Mol Oncol 2008;2(4)296-316.

[92] Nimonkar AV, Ozsoy AZ, Genschel J, Modrich P, Kowalczykowski SC. Human exo‐nuclease 1 and BLM helicase interact to resect DNA and initiate DNA repair. ProcNatl Acad Sci U S A 2008;105(44) 16906-16911.

[93] German J. Bloom's syndrome. XX. The first 100 cancers. Cancer Genet Cytogenet1997;93(1) 100-106.

[94] Bohr VA. Deficient DNA repair in the human progeroid disorder, Werner syndrome.Mutat Res 2005;577(1-2) 252-259.

[95] Monnat RJ Jr. Human RECQ helicases: roles in DNA metabolism, mutagenesis andcancer biology. Semin Cancer Biol 2010;20(5) 329-339.

[96] Singh DK, Ahn B, Bohr VA. Roles of RECQ helicases in recombination based DNArepair, genomic stability and aging. Biogerontology 2009;10(3) 235-252.

[97] Anbari KK, Ierardi-Curto LA, Silber JS, Asada N, Spinner N, Zackai EH, Belasco J,Morrissette JD, Dormans JP. Two primary osteosarcomas in a patient with Roth‐mund-Thomson syndrome. Clin Orthop Relat Res 2000;(378) 213-223.

[98] Thompson LH, Hinz JM. Cellular and molecular consequences of defective Fanconianemia proteins in replication-coupled DNA repair: mechanistic insights. Mutat Res2009;668(1-2) 54-72.

[99] Alter BP. Cancer in Fanconi anemia, 1927-2001. Cancer 2003;97(2) 425-440.

[100] Lehmann AR, McGibbon D, Stefanini M. Xeroderma pigmentosum. Orphanet J RareDis 2011;6 70.

[101] Oh KS, Imoto K, Emmert S, Tamura D, DiGiovanna JJ, Kraemer KH. Nucleotide exci‐sion repair proteins rapidly accumulate but fail to persist in human XP-E (DDB2 mu‐tant) cells. Photochem Photobiol 2011;87(3) 729-733.

[102] Manchanda R, Menon U, Michaelson-Cohen R, Beller U, Jacobs I. Hereditary non-polyposis colorectal cancer or Lynch syndrome: the gynaecological perspective. CurrOpin Obstet Gynecol 2009;21(1) 31-38.

[103] David SS, O'Shea VL, Kundu S. Base-excision repair of oxidative DNA damage. Na‐ture 2007;447(7147) 941-950.

[104] Cleary SP, Cotterchio M, Jenkins MA, Kim H, Bristow R, Green R, Haile R, HopperJL, LeMarchand L, Lindor N, Parfrey P, Potter J, Younghusband B, Gallinger S.

DNA Damage, DNA Repair and Cancerhttp://dx.doi.org/10.5772/53919

455

Page 44: DNA Damage, DNA Repair and Cancer - InTech - Opencdn.intechopen.com/pdfs/43929/InTech-Dna_damage... · Chapter 16 DNA Damage, DNA Repair and Cancer Carol Bernstein, Anil R. Prasad,

Germline MutY human homologue mutations and colorectal cancer: a multisite case-control study. Gastroenterology 2009;136(4) 1251-1260.

[105] López-Contreras AJ, Fernandez-Capetillo O. The ATR barrier to replication-bornDNA damage. DNA Repair (Amst) 2010;9(12) 1249-1255.

[106] Tanaka A, Weinel S, Nagy N, O'Driscoll M, Lai-Cheong JE, Kulp-Shorten CL, KnableA, Carpenter G, Fisher SA, Hiragun M, Yanase Y, Hide M, Callen J, McGrath JA.Germline mutation in ATR in autosomal- dominant oropharyngeal cancer syndrome.Am J Hum Genet 2012;90(3) 511-517.

[107] Viktorsson K, De Petris L, Lewensohn R. The role of p53 in treatment responses oflung cancer. Biochem Biophys Res Commun 2005;331(3) 868-880.

[108] Esteller M, Silva JM, Dominguez G, Bonilla F, Matias-Guiu X, Lerma E, Bussaglia E,Prat J, Harkes IC, Repasky EA, Gabrielson E, Schutte M, Baylin SB, Herman JG. Pro‐moter hypermethylation and BRCA1 inactivation in sporadic breast and ovarian tu‐mors. J Natl Cancer Inst 2000;92(7) 564-569.

[109] Shen L, Kondo Y, Rosner GL, Xiao L, Hernandez NS, Vilaythong J, Houlihan PS,Krouse RS, Prasad AR, Einspahr JG, Buckmeier J, Alberts DS, Hamilton SR, Issa JP.MGMT promoter methylation and field defect in sporadic colorectal cancer. J NatlCancer Inst 2005;97(18) 1330-1338.

[110] Psofaki V, Kalogera C, Tzambouras N, Stephanou D, Tsianos E, Seferiadis K, KoliosG. Promoter methylation status of hMLH1, MGMT, and CDKN2A/p16 in colorectaladenomas. World J Gastroenterol 2010;16(28) 3553-3560.

[111] Lee KH, Lee JS, Nam JH, Choi C, Lee MC, Park CS, Juhng SW, Lee JH. Promotermethylation status of hMLH1, hMSH2, and MGMT genes in colorectal cancer associ‐ated with adenoma-carcinoma sequence. Langenbecks Arch Surg 2011;396(7)1017-1026.

[112] Facista A, Nguyen H, Lewis C, Prasad AR, Ramsey L, Zaitlin B, Nfonsam V, KrouseRS, Bernstein H, Payne CM, Stern S, Oatman N, Banerjee B, Bernstein C. Deficient ex‐pression of DNA repair enzymes in early progression to sporadic colon cancer. Ge‐nome Integr 2012;3(1): 3.

[113] Zou XP, Zhang B, Zhang XQ, Chen M, Cao J, Liu WJ. Promoter hypermethylation ofmultiple genes in early gastric adenocarcinoma and precancerous lesions. Hum Path‐ol 2009;40(11) 1534-1542.

[114] Ling ZQ, Li P, Ge MH, Hu FJ, Fang XH, Dong ZM, Mao WM. Aberrant methylationof different DNA repair genes demonstrates distinct prognostic value for esophagealcancer. Dig Dis Sci 2011;56(10) 2992-3004.

[115] Lee KH, Lee JS, Nam JH, Choi C, Lee MC, Park CS, Juhng SW, Lee JH. Promotermethylation status of hMLH1, hMSH2, and MGMT genes in colorectal cancer associ‐ated with adenoma-carcinoma sequence. Langenbecks Arch Surg 2011;396(7)1017-1026.

New Research Directions in DNA Repair456

Page 45: DNA Damage, DNA Repair and Cancer - InTech - Opencdn.intechopen.com/pdfs/43929/InTech-Dna_damage... · Chapter 16 DNA Damage, DNA Repair and Cancer Carol Bernstein, Anil R. Prasad,

[116] Paluszczak J, Misiak P, Wierzbicka M, Woźniak A, Baer-Dubowska W. Frequent hy‐permethylation of DAPK, RARbeta, MGMT, RASSF1A and FHIT in laryngeal squa‐mous cell carcinomas and adjacent normal mucosa. Oral Oncol 2011;47(2) 104-107.

[117] Guo M, House MG, Hooker C, Han Y, Heath E, Gabrielson E, Yang SC, Baylin SB,Herman JG, Brock MV. Promoter hypermethylation of resected bronchial margins: afield defect of changes? Clin Cancer Res 2004;10(15) 5131-5136.

[118] Skiriute D, Vaitkiene P, Saferis V, Asmoniene V, Skauminas K, Deltuva VP, Tama‐sauskas A. MGMT, GATA6, CD81, DR4, and CASP8 gene promoter methylation inglioblastoma. BMC Cancer 2012;12(1) 218.

[119] Pogribny IP, James SJ. Reduction of p53 gene expression in human primary hepato‐cellular carcinoma is associated with promoter region methylation without codingregion mutation. Cancer Lett 2002;176(2) 169-74.

[120] Guan H, Ji M, Hou P, Liu Z, Wang C, Shan Z, Teng W, Xing M. Hypermethylation ofthe DNA mismatch repair gene hMLH1 and its association with lymph node meta‐stasis and T1799A BRAF mutation in patients with papillary thyroid cancer. Cancer2008;113(2) 247-255.

[121] Wan G, Mathur R, Hu X, Zhang X, Lu X. miRNA response to DNA damage. TrendsBiochem Sci 2011;36(9) 478-484.

[122] Malumbres M. miRNAs and cancer: An epigenetics view. Mol Aspects Med 2012 Jul4 [Epub ahead of print]

[123] Resar LM. The high mobility group A1 gene: transforming inflammatory signals intocancer? Cancer Res 2010;70(2) 436-439.

[124] Baldassarre G, Battista S, Belletti B, Thakur S, Pentimalli F, Trapasso F, Fedele M,Pierantoni G, Croce CM, Fusco A. Negative regulation of BRCA1 gene expression byHMGA1 proteins accounts for the reduced BRCA1 protein levels in sporadic breastcarcinoma. Mol Cell Biol 2003;23(7) 2225-2238.

[125] Borrmann L, Schwanbeck R, Heyduk T, Seebeck B, Rogalla P, Bullerdiek J, Wisniew‐ski JR. High mobility group A2 protein and its derivatives bind a specific region ofthe promoter of DNA repair gene ERCC1 and modulate its activity. Nucleic AcidsRes 2003;31(23) 6841-6851.

[126] Lee S, Jung JW, Park SB, Roh K, Lee SY, Kim JH, Kang SK, Kang KS. Histone deacety‐lase regulates high mobility group A2-targeting microRNAs in human cord blood-derived multipotent stem cell aging. Cell Mol Life Sci 2011;68(2) 325-336.

[127] Palmieri D, D'Angelo D, Valentino T, De Martino I, Ferraro A, Wierinckx A, FedeleM, Trouillas J, Fusco A. Downregulation of HMGA-targeting microRNAs has a criti‐cal role in human pituitary tumorigenesis. Oncogene 2011 Dec 5. doi: 10.1038/onc.2011.557.

DNA Damage, DNA Repair and Cancerhttp://dx.doi.org/10.5772/53919

457

Page 46: DNA Damage, DNA Repair and Cancer - InTech - Opencdn.intechopen.com/pdfs/43929/InTech-Dna_damage... · Chapter 16 DNA Damage, DNA Repair and Cancer Carol Bernstein, Anil R. Prasad,

[128] Sampath D, Liu C, Vasan K, Sulda M, Puduvalli VK, Wierda WG, Keating MJ. His‐tone deacetylases mediate the silencing of miR-15a, miR-16, and miR-29b in chroniclymphocytic leukemia. Blood 2012;119(5) 1162-1172.

[129] D'Angelo D, Palmieri D, Mussnich P, Roche M, Wierinckx A, Raverot G, Fedele M,Croce CM, Trouillas J, Fusco A. Altered MicroRNA Expression Profile in Human Pi‐tuitary GH Adenomas: Down-Regulation of miRNA Targeting HMGA1, HMGA2,and E2F1. J Clin Endocrinol Metab 2012;97(7) E1128-1138.

[130] Suzuki H, Takatsuka S, Akashi H, Yamamoto E, Nojima M, Maruyama R, Kai M, Ya‐mano HO, Sasaki Y, Tokino T, Shinomura Y, Imai K, Toyota M. Genome-wide profil‐ing of chromatin signatures reveals epigenetic regulation of MicroRNA genes incolorectal cancer. Cancer Res 2011;71(17) 5646-5658.

[131] Tchou-Wong KM, Kiok K, Tang Z, Kluz T, Arita A, Smith PR, Brown S, Costa M. Ef‐fects of nickel treatment on H3K4 trimethylation and gene expression. PLoS One2011;6(3) e17728.

[132] Klase Z, Winograd R, Davis J, Carpio L, Hildreth R, Heydarian M, Fu S, McCaffrey T,Meiri E, Ayash-Rashkovsky M, Gilad S, Bentwich Z, Kashanchi F: HIV-1 TAR miR‐NA protects against apoptosis by altering cellular gene expression. Retrovirology2009;6 18.

[133] http://www.ebi.ac.uk/enright-srv/microcosm/htdocs/targets/v5/

[134] Wang P, Dong Q, Zhang C, Kuan PF, Liu Y, Jeck WR, Andersen JB, Jiang W, SavichGL, Tan TX, Auman JT, Hoskins JM, Misher AD, Moser CD, Yourstone SM, Kim JW,Cibulskis K, Getz G, Hunt HV, Thorgeirsson SS, Roberts LR, Ye D, Guan KL, XiongY, Qin LX, Chiang DY. Mutations in isocitrate dehydrogenase 1 and 2 occur frequent‐ly in intrahepatic cholangiocarcinomas and share hypermethylation targets with glio‐blastomas. Oncogene 2012 Jul 23. doi: 10.1038/onc.2012.315. [Epub ahead of print]

[135] Watanabe T, Nobusawa S, Kleihues P, Ohgaki H. IDH1 mutations are early events inthe development of astrocytomas and oligodendrogliomas. Am J Pathol 2009;174(4)1149-1153.

[136] Turcan S, Rohle D, Goenka A, Walsh LA, Fang F, Yilmaz E, Campos C, Fabius AW,Lu C, Ward PS, Thompson CB, Kaufman A, Guryanova O, Levine R, Heguy A, VialeA, Morris LG, Huse JT, Mellinghoff IK, Chan TA. IDH1 mutation is sufficient to es‐tablish the glioma hypermethylator phenotype. Nature 2012;483(7390) 479-483.

[137] Carrillo JA, Lai A, Nghiemphu PL, Kim HJ, Phillips HS, Kharbanda S, Moftakhar P,Lalaezari S, Yong W, Ellingson BM, Cloughesy TF, Pope WB. Relationship betweenTumor Enhancement, Edema, IDH1 Mutational Status, MGMT Promoter Methyla‐tion, and Survival in Glioblastoma. AJNR Am J Neuroradiol 2012;33(7) 1349-1355.

[138] Nelson PT, Kiriakidou M, Mourelatos Z, Tan GS, Jennings MH, Xie K, Wang WX.High-throughput experimental studies to identify miRNA targets directly, with spe‐cial focus on the mammalian brain. Brain Res 2010;1338 122-130.

New Research Directions in DNA Repair458

Page 47: DNA Damage, DNA Repair and Cancer - InTech - Opencdn.intechopen.com/pdfs/43929/InTech-Dna_damage... · Chapter 16 DNA Damage, DNA Repair and Cancer Carol Bernstein, Anil R. Prasad,

[139] Ohka F, Natsume A, Motomura K, Kishida Y, Kondo Y, Abe T, Nakasu Y, Namba H,Wakai K, Fukui T, Momota H, Iwami K, Kinjo S, Ito M, Fujii M, Wakabayashi T. Theglobal DNA methylation surrogate LINE-1 methylation is correlated with MGMTpromoter methylation and is a better prognostic factor for glioma. PLoS One2011;6(8) e23332.

[140] Bernstein C, Bernstein H, Payne CM, Dvorak K, Garewal H. Field defects in progres‐sion to gastrointestinal tract cancers. Cancer Lett 2008;260(1-2) 1-10.

[141] Gerlinger M, Rowan AJ, Horswell S, Larkin J, Endesfelder D, Gronroos E, MartinezP, Matthews N, Stewart A, Tarpey P, Varela I, Phillimore B, Begum S, McDonald NQ,Butler A, Jones D, Raine K, Latimer C, Santos CR, Nohadani M, Eklund AC, Spencer-Dene B, Clark G, Pickering L, Stamp G, Gore M, Szallasi Z, Downward J, Futreal PA,Swanton C. N Engl J Med 2012;366(10) 883-892.

[142] Bernstein C, Facista A, Nguyen H, Zaitlin B, Hassounah N, Loustaunau C, PayneCM, Banerjee B, Goldschmid S, Tsikitis VL, Krouse R, Bernstein H. Cancer and agerelated colonic crypt deficiencies in cytochrome c oxidase I. World J Gastrointest On‐col 2010;2(12) 429-442.

[143] Nicolas P, Kim KM, Shibata D, Tavaré S: The stem cell population of the human co‐lon crypt: analysis via methylation patterns. PLoS Comput Biol 2007;3 e28.

[144] Willis ND, Przyborski SA, Hutchison CJ, Wilson RG: Colonic and colorectal cancerstem cells: progress in the search for putative biomarkers. J Anat 2008;213 59-65.

[145] Loeb LA. Human cancers express mutator phenotypes: origin, consequences and tar‐geting. Nat Rev Cancer 2011;11(6) 450-457.

[146] Roy M, Sinha D, Mukherjee S, Biswas J. Curcumin prevents DNA damage and en‐hances the repair potential in a chronically arsenic-exposed human population inWest Bengal, India. Eur J Cancer Prev 2011;20(2) 123-131.

[147] Qin XJ, Liu W, Li YN, Sun X, Hai CX, Hudson LG, Liu KJ. Poly(ADP-ribose) poly‐merase-1 inhibition by arsenite promotes the survival of cells with unrepaired DNAlesions induced by UV exposure. Toxicol Sci 2012;127(1) 120-129.

[148] Ebert F, Weiss A, Bültemeyer M, Hamann I, Hartwig A, Schwerdtle T. Arsenicals af‐fect base excision repair by several mechanisms. Mutat Res 2011;715(1-2) 32-41.

[149] Andrew AS, Karagas MR, Hamilton JW. Decreased DNA repair gene expressionamong individuals exposed to arsenic in United States drinking water. Int J Cancer2003;104(3) 263-268.

[150] Nollen M, Ebert F, Moser J, Mullenders LH, Hartwig A, Schwerdtle T. Impact of ar‐senic on nucleotide excision repair: XPC function, protein level, and gene expression.Mol Nutr Food Res 2009;53(5) 572-582.

DNA Damage, DNA Repair and Cancerhttp://dx.doi.org/10.5772/53919

459

Page 48: DNA Damage, DNA Repair and Cancer - InTech - Opencdn.intechopen.com/pdfs/43929/InTech-Dna_damage... · Chapter 16 DNA Damage, DNA Repair and Cancer Carol Bernstein, Anil R. Prasad,

[151] Shen S, Lee J, Weinfeld M, Le XC. Attenuation of DNA damage-induced p53 expres‐sion by arsenic: a possible mechanism for arsenic co-carcinogenesis. Mol Carcinog2008;47(7) 508-518.

[152] Zhou ZH, Lei YX, Wang CX. Analysis of aberrant methylation in DNA repair genesduring malignant transformation of human bronchial epithelial cells induced by cad‐mium. Toxicol Sci 2012;125(2) 412-417.

[153] Wieland M, Levin MK, Hingorani KS, Biro FN, Hingorani MM. Mechanism of cadmi‐um-mediated inhibition of Msh2-Msh6 function in DNA mismatch repair. Biochem‐istry 2009;48(40) 9492-9502.

[154] Bravard A, Vacher M, Gouget B, Coutant A, de Boisferon FH, Marsin S, Chevillard S,Radicella JP. Redox regulation of human OGG1 activity in response to cellular oxida‐tive stress. Mol Cell Biol 2006;26(20) 7430-7436.

[155] Viau M, Gastaldo J, Bencokova Z, Joubert A, Foray N. Cadmium inhibits non-homol‐ogous end-joining and over-activates the MRE11-dependent repair pathway. MutatRes 2008;654(1) 13-21.

[156] Schwerdtle T, Ebert F, Thuy C, Richter C, Mullenders LH, Hartwig A. Genotoxicityof soluble and particulate cadmium compounds: impact on oxidative DNA damageand nucleotide excision repair. Chem Res Toxicol 2010;23(2) 432-442.

[157] Burnat G, Majka J, Konturek PC. Bile acids are multifunctional modulators of theBarrett's carcinogenesis. J Physiol Pharmacol 2010;61(2) 185-192.

[158] Romagnolo DF, Chirnomas RB, Ku J, Jeffy BD, Payne CM, Holubec H, Ramsey L,Bernstein H, Bernstein C, Kunke K, Bhattacharyya A, Warneke J, Garewal H. Deoxy‐cholate, an endogenous tumor promoter and DNA damaging agent, modulatesBRCA-1 expression in apoptosis-sensitive epithelial cells: loss of BRCA-1 expressionin colonic adenocarcinomas. Nutr Cancer 2003;46(1) 82-92.

[159] Ogawa A, Murate T, Suzuki M, Nimura Y, Yoshida S. Lithocholic acid, a putative tu‐mor promoter, inhibits mammalian DNA polymerase beta. Jpn J Cancer Res1998;89(11) 1154-1159.

[160] Feng Z, Hu W, Tang MS. Trans-4-hydroxy-2-nonenal inhibits nucleotide excision re‐pair in human cells: a possible mechanism for lipid peroxidation-induced carcino‐genesis. Proc Natl Acad Sci U S A 2004;101(23) 8598-8602.

[161] Feng Z, Hu W, Marnett LJ, Tang MS. Malondialdehyde, a major endogenous lipidperoxidation product, sensitizes human cells to UV- and BPDE-induced killing andmutagenesis through inhibition of nucleotide excision repair. Mutat Res2006;601(1-2) 125-136.

[162] Chang CL, Marra G, Chauhan DP, Ha HT, Chang DK, Ricciardiello L, Randolph A,Carethers JM, Boland CR. Oxidative stress inactivates the human DNA mismatch re‐pair system. Am J Physiol Cell Physiol 2002;283(1) C148-154.

New Research Directions in DNA Repair460

Page 49: DNA Damage, DNA Repair and Cancer - InTech - Opencdn.intechopen.com/pdfs/43929/InTech-Dna_damage... · Chapter 16 DNA Damage, DNA Repair and Cancer Carol Bernstein, Anil R. Prasad,

[163] Langie SA, Knaapen AM, Houben JM, van Kempen FC, de Hoon JP, Gottschalk RW,Godschalk RW, van Schooten FJ. The role of glutathione in the regulation of nucleo‐tide excision repair during oxidative stress. Toxicol Lett 20075;168(3) 302-309.

[164] Hill JW, Hu JJ, Evans MK. OGG1 is degraded by calpain following oxidative stressand cisplatin exposure. DNA Repair (Amst) 2008;7(4) 648-654.

[165] Sudprasert W, Navasumrit P, Ruchirawat M. Effects of low-dose gamma radiationon DNA damage, chromosomal aberration and expression of repair genes in humanblood cells. Int J Hyg Environ Health. 2006;209(6) 503-511.

[166] Li D, Wang Q, Liu C, Duan H, Zeng X, Zhang B, Li X, Zhao J, Tang S, Li Z, Xing X,Yang P, Chen L, Zeng J, Zhu X, Zhang S, Zhang Z, Ma L, He Z, Wang E, Xiao Y,Zheng Y, Chen W. Aberrant expression of miR-638 contributes to benzo(a)pyrene-in‐duced human cell transformation. Toxicol Sci 2012;125(2) 382-391.

[167] Liu WB, Ao L, Cui ZH, Zhou ZY, Zhou YH, Yuan XY, Xiang YL, Cao J, Liu JY. Molec‐ular analysis of DNA repair gene methylation and protein expression during chemi‐cal-induced rat lung carcinogenesis. Biochem Biophys Res Commun 2011;408(4)595-601.

[168] Hanova M, Stetina R, Vodickova L, Vaclavikova R, Hlavac P, Smerhovsky Z, Naccar‐ati A, Polakova V, Soucek P, Kuricova M, Manini P, Kumar R, Hemminki K, VodickaP. Modulation of DNA repair capacity and mRNA expression levels of XRCC1,hOGG1 and XPC genes in styrene-exposed workers. Toxicol Appl Pharmacol2010;248(3) 194-200.

[169] Chen YY, Chung JG, Wu HC, Bau DT, Wu KY, Kao ST, Hsiang CY, Ho TY, ChiangSY. Aristolochic acid suppresses DNA repair and triggers oxidative DNA damage inhuman kidney proximal tubular cells. Oncol Rep 2010;24(1) 141-153.

[170] Grosskopf C, Schwerdtle T, Mullenders LH, Hartwig A. Antimony impairs nucleo‐tide excision repair: XPA and XPE as potential molecular targets. Chem Res Toxicol2010;23(7) 1175-1183.

[171] Ji W, Yang L, Yu L, Yuan J, Hu D, Zhang W, Yang J, Pang Y, Li W, Lu J, Fu J, Chen J,Lin Z, Chen W, Zhuang Z. Epigenetic silencing of O6-methylguanine DNA methyl‐transferase gene in NiS-transformed cells. Carcinogenesis 2008;29(6) 1267-1275.

[172] Link A, Balaguer F, Goel A. Cancer chemoprevention by dietary polyphenols: prom‐ising role for epigenetics. Biochem Pharmacol 2010;80(12) 1771-1792.

[173] Fang M, Chen D, Yang CS. Dietary polyphenols may affect DNA methylation. J Nutr2007;137(1 Suppl) 223S-228S.

[174] Olaharski AJ, Rine J, Marshall BL, Babiarz J, Zhang L, Verdin E, Smith MT. The fla‐voring agent dihydrocoumarin reverses epigenetic silencing and inhibits sirtuin de‐acetylases. PLoS Genet 2005;1(6) e77.

DNA Damage, DNA Repair and Cancerhttp://dx.doi.org/10.5772/53919

461

Page 50: DNA Damage, DNA Repair and Cancer - InTech - Opencdn.intechopen.com/pdfs/43929/InTech-Dna_damage... · Chapter 16 DNA Damage, DNA Repair and Cancer Carol Bernstein, Anil R. Prasad,

[175] Kikuno N, Shiina H, Urakami S, Kawamoto K, Hirata H, Tanaka Y, Majid S, IgawaM, Dahiya R. Genistein mediated histone acetylation and demethylation activates tu‐mor suppressor genes in prostate cancer cells. Int J Cancer 2008;123(3) 552-560.

[176] Collins AR, Azqueta A, Langie SA. Effects of micronutrients on DNA repair. Eur JNutr 2012;51(3) 261-279.

[177] http://www.phenol-explorer.eu

[178] http://www.ajcn.org/content/93/6/1220.full.pdf+html

[179] http://www.hmdb.ca/search/search?query=caffeic+acid

[180] Aiyer HS, Vadhanam MV, Stoyanova R, Caprio GD, Clapper ML, Gupta RC. Dietaryberries and ellagic acid prevent oxidative DNA damage and modulate expression ofDNA repair genes. Int J Mol Sci 2008;9(3) 327-341.

[181] Niture SK, Velu CS, Smith QR, Bhat GJ, Srivenugopal KS. Increased expression of theMGMT repair protein mediated by cysteine prodrugs and chemopreventative natu‐ral products in human lymphocytes and tumor cell lines. Carcinogenesis 2007;28(2)378-389.

[182] Bernstein H, Crowley-Skillicorn C, Bernstein C, Payne CM, Dvorak K, Garewal H.Dietary Compounds that Enhance DNA Repair and their Relevance to Cancer andAging. In: BR Landseer (ed.) New Research on DNA Repair. New York: Nova Sci‐ence Publishers, Inc.; 2007. p99-113.

[183] Clifford MN. Chlorogenic acids and other cinnamates --- nature, occurrence and diet‐ary burden. J Sci Food Agric 1999;79(3) 362-372.

[184] Mattila P, Kumpulainen J. Determination of free and total phenolic acids in plant-de‐rived foods by HPLC with diode-array detection. J Agric Food Chem 2002;50(13)3660-3667.

[185] Yun MH, Hiom K. Understanding the functions of BRCA1 in the DNA-damage re‐sponse. Biochem Soc Trans 2009;37(Pt 3) 597-604.

[186] Quinn JE, Kennedy RD, Mullan PB, Gilmore PM, Carty M, Johnston PG, Harkin DP.BRCA1 functions as a differential modulator of chemotherapy-induced apoptosis.Cancer Res 2003;63(19) 6221-6228.

[187] Taron M, Rosell R, Felip E, Mendez P, Souglakos J, Ronco MS, Queralt C, Majo J, San‐chez JM, Sanchez JJ, Maesire J. BRCA1 mRNA expression levels as an indicator ofchemoresistance in lung cancer. Human Molecular Genetics 2004;13(20) 2443-2449.

[188] Papadaki C, Sfakianaki M, Ioannidis G, Lagoudaki E, Trypaki M, Tryfonidis K, Mav‐roudis D, Stathopoulos E, Georgoulias V, Souglakos J. ERCC1 and BRCA1 mRNA ex‐pression levels in the primary tumor could predict the effectiveness of the second-line cisplatin-based chemotherapy in pretreated patents with metastatic non-smallcell lung cancer. J Thorac Oncol 2012;7(4) 663-671.

New Research Directions in DNA Repair462

Page 51: DNA Damage, DNA Repair and Cancer - InTech - Opencdn.intechopen.com/pdfs/43929/InTech-Dna_damage... · Chapter 16 DNA Damage, DNA Repair and Cancer Carol Bernstein, Anil R. Prasad,

[189] Weberpals J, Garbuio K, O’Brien A, Clark-Knowles K, Doucette S, Antoniouk O,Goss G, Dimitroulakos J. The DNA repair proteins BRCA1 and ERCC1 as predictivemarkers in sporadic ovarian cancer. Int J Cancer 2009;124(4) 806-815.

[190] Reed E. Platinum-DNA adduct, nucleotide excision repair and platinum based anti-cancer chemotherapy. Cancer Treat Rev 1998;24(5) 331-344.

[191] Kelland L. The resurgence of platinum-based cancer chemotherapy. Nat Rev Cancer2007;7(8) 573-584.

[192] Martin LP, Hamilton TC, Schilder RJ. Platinum Resistance: The role of DNA repairpathways. Clin Cancer Res 2008;14(5) 1291-1295.

[193] Bosl GJ, Motzer RJ. Testicular germ-cell cancer. N Engl J Med 1997;337(4) 242-253.

[194] Welsh C, Day R, McGurk C, Masters JRW, Wood RD, Koberle B. Reduced levels ofXPA, ERCC1 and XPF DNA repair proteins in testis tumor cell lines. Int J Cancer2004;110(3) 352-361.

[195] Simon GR, Sharma S, Cantor A, Smith P, Bepler G. ERCC1 expression is a predictorof survival in resected patients with non-small cell lung cancer. Chest 2005;127(3)978-983.

[196] Lord RVN, Brabender J, Gandara D, Alberola V, Camps C, Domine M, Cardenal F,Sanchez JM, Gumerlock PH, Taron M, Sanchez JJ, Danenberg KD, Danenberg PV,Rosell R. Low ERCC1 expression correlates with prolonged survival after cisplatinplus gemcitabine chemotherapy in non-small cell lung cancer. Clin Cancer Res2002;8(7) 2286-2291.

[197] Zhou W, Gurubhagavatula S, Liu G, Park S, Neuberg DS, Wain JC, Lynch TJ, Su L,Christiani DC. Excision repair cross-complementation group 1 polymorphism pre‐dicts overall survival in advanced non-small cell lung cancer patients treated withplatinum-based chemotherapy. Clin Cancer Res 2004;10(15) 4939-4943.

[198] Olaussen KA, Dunant A, Fouret P, Brambilla E, Andre F, Haddad V, Taranchon E,Filipits M, Pirker R, Popper HH, Stahel R, Sabatier L, Pignon J-P, Tursz T, Le Chava‐lier T, Soria, J-C. DNA repair by ERCC1 in non-small-cell lung cancer and cisplatin-based adjuvant chemotherapy. N Engl J Med 2006;355(10) 983-991.

[199] Leng XF, Chen MW, Xian L, Dai L, Ma GY, Li MH. Combined analysis of mRNA ex‐pression of ERCC1, BAG-1, BRCA1, RRM1 and TUBB3 to predict prognosis in pa‐tients with non-small cell lung cancer who received adjuvant chemotherapy. J ExpClin Cancer Res 2012;23 31:25.

[200] Dabholkar M, Bostick-Bruton F, Weber C, Bohr VA, Egwuagu C, Reed E. ERCC1 andERCC2 expression in malignant tissues from ovarian cancer patients. J Natl CancerInst 1992;84(19) 1512-1517.

[201] Kang S, Ju W, Kim JW, Park N-H, Song Y-S, Kim SC, Park S-Y, Kang S-B, Lee H-P.Association between excision repair cross-complementation group 1 polymorphism

DNA Damage, DNA Repair and Cancerhttp://dx.doi.org/10.5772/53919

463

Page 52: DNA Damage, DNA Repair and Cancer - InTech - Opencdn.intechopen.com/pdfs/43929/InTech-Dna_damage... · Chapter 16 DNA Damage, DNA Repair and Cancer Carol Bernstein, Anil R. Prasad,

and clinical outcome of platinum based chemotherapy in patients with epithelialovarian cancer. Exp Mol Med 2006;8(3) 320-324.

[202] Shirota Y, Stoehlmacher J, Brabender J, Xiong Y-P, Uetake H, Danenberg KD, Grosh‐en S, Tsao-Wei DD, Danenberg PV, Lenz H-J. ERCC1 and thymidylate synthasemRNA levels predict survival for colorectal cancer patients receiving combinationoxaliplatin and fluorouracil chemotherapy. J Clin Oncol 2001;19(23) 4298-4304.

[203] Viguier J, Boige V, Miquel C, Pocard M, Giraudeau B, Sabourin J-C, Ducreux M, Sar‐asin A, Praz F. ERCC1 codon 118 polymorphism is a predictive factor for the tumorresponse to oxaliplatin/5-fluorouracil combination chemotherapy in patients with ad‐vanced colorectal cancer. Clin Cancer Res 2005;11(17) 6212-6217.

[204] Metzger R, Leichman CG, Danenberg KD, Danenberg PV, Lenz H-J, Hayashi K,Groshen S, Salonga D, Cohen H, Laine L, Crookes P, Silberman H, Barando J, KondaB, Leichman L. ERCC1 mRNA levels complement thymidylate synthase mRNA lev‐els in predicting response and survival for gastric cancer patients receiving combina‐tion cisplatin and fluorouracil chemotherapy. J Clin Oncol 1998;16(1) 309-316.

[205] Bellmunt J, Paz-Ares L, Cuello M, Cecere FL, Albiol S, Guillem V, Gallardo E, CarlesJ, Mendez P, de la Cruz JJ, Taron M, Rosell R, Baselga J; Spanish Oncology Genitouri‐nary Group. Gene expression of ERCC1 as a novel prognostic marker in advancedbladder cancer patients receiving cisplatin-based chemotherapy. Ann Oncol2007;18(3) 522-528.

[206] Esteller M, Garcia-Foncillas J, Andion E, Goodman SN, Hidalgo OF, Vanaclocha V,Baylin SB, Herman JG. Inactivation of the DNA-repair gene MGMT and the clinicalresponse of gliomas to alkylating agents. N Engl J Med 2000;343(19) 1350-1354.

[207] Paz MF, Yaya-Tur R, Rojas-Marcos I, Reynes G, Pollan M, Aguirre-Cruz L, Garcia-Lopez JL, Piquer J, Safont M-J, Balana C, Sanchez-Cespedes M, Garcia-Villanueva M,Arribas L, Esteller M. CpG island hypermethylation of the DNA repair enzymemethyltransferase predicts response to temozolomide in primary gliomas. Clin Can‐cer Res 2004;10(15) 4933-4938.

[208] Hegi ME, Diserens A-C, Godard S, Dietrich P-Y, Regli L, Ostermann S, Otten P, VanMelle G, de Tribolet N, Stupp R. Clinical trial substantiates the predictive value ofO-6-methylguanine-DNA methyltransferase promoter methylation in glioblastomapatients treated with temozolomide. Clin Cancer Res 2004;10(6) 1871-1874.

[209] Hegi ME, Liu L, Herman JG, Stupp R, Wick W, Weller M, Mehta MP, Gilbert MR.Correlation of O6-methylguanine methyltransferase (MGMT) promoter methylationwith clinical outcomes in glioblastoma and clinical strategies to modulate MGMT ac‐tivity. J Clin Oncol 2008;26(25) 4189-4199.

[210] Weller M, Stupp R, Reifenberger G, Brandes AA, van den Bent MJ, Wick W, HegiME. MGMT promoter methylation in malignant gliomas: ready for personalizedmedicine? Nat Rev Neurol 2010;6(1) 39-51.

New Research Directions in DNA Repair464

Page 53: DNA Damage, DNA Repair and Cancer - InTech - Opencdn.intechopen.com/pdfs/43929/InTech-Dna_damage... · Chapter 16 DNA Damage, DNA Repair and Cancer Carol Bernstein, Anil R. Prasad,

[211] Leung M, Rosen D, Fields S, Cesano A, Budman DR. Poly(ADP-ribose) polymerase-1inhibition: preclinical and clinical development of synthetic lethality. Mol Med2011;17(7-8) 854-862. doi: 10.2119/molmed.2010.00240.

[212] Basu B, Yap TA, Molife LR, de Bono JS. Targeting the DNA damage response in on‐cology: past, present and future perspectives. Curr Opin Oncol 2012;24(3) 316-324.

[213] Fong PC, Boss DS, Yap TA, Tutt A, Wu P, Roelvink, MM, Mortimer P, Swaisland H,Lau A, O’Connor MJ, Ashworth A, Carmichael J, Kaye SB, Schellens JHM, de BonoJS. Inhibition of poly(ADP-ribose) polymerase in tumors from BRCA mutation carri‐ers. N Engl J Med 2009;361(2) 123-134.

[214] Tutt A, Robson M, Garber JE, Domchek SM, Audeh MW, Weitzel JN, Friedlander M,Arun B, Loman N, Schmutzler RK, Wardley A, Mitchell G, Earl H, Wickens M, Car‐michael J. Oral poly(ADP-ribose) polymerase inhibitor olaparib in patients withBRCA1 or BRCA2 mutations and advanced breast cancer: a proof-of-concept trial.Lancet 2010;376(9737) 235-244.

[215] Audeh MW, Carmichael J, Penson RT, Friedlander M, Powell B, Bell-McGuinn KM,Scott C, Weitzel JN, Oaknin A, Loman N, Lu K, Schmutzler RK, Matulonis U, Wick‐ens M, Tutt A. Oral poly(ADP-ribose) polymerase inhibitor olaparib in patients withBRCA1 or BRCA2 mutations and recurrent ovarian cancer: a proof-of-concept trial.Lancet 2010;376(9737) 245-251.

[216] Stratton MR. Exploring the genomes of cancer cells: progress and promise. Science2011;331(6024) 1553-1558.

[217] Hanahan D, Weinberg RA. Hallmarks of cancer: the next generation. Cell 2011;144(5)646-674.

[218] Lord CJ, Ashworth A. The DNA damage response and cancer therapy. Nature2012;481(7382) 287-294.

DNA Damage, DNA Repair and Cancerhttp://dx.doi.org/10.5772/53919

465

Page 54: DNA Damage, DNA Repair and Cancer - InTech - Opencdn.intechopen.com/pdfs/43929/InTech-Dna_damage... · Chapter 16 DNA Damage, DNA Repair and Cancer Carol Bernstein, Anil R. Prasad,